Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 151
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Cell Dev Biol ; 31: 125-47, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26436705

RESUMEN

Hepatitis B virus (HBV) infection affects 240 million people worldwide. A liver-specific bile acid transporter named the sodium taurocholate cotransporting polypeptide (NTCP) has been identified as the cellular receptor for HBV and its satellite, the hepatitis D virus (HDV). NTCP likely acts as a major determinant for the liver tropism and species specificity of HBV and HDV at the entry level. NTCP-mediated HBV entry interferes with bile acid transport in cell cultures and has been linked with alterations in bile acid and cholesterol metabolism in vivo. The human liver carcinoma cell line HepG2, complemented with NTCP, now provides a valuable platform for studying the basic biology of the viruses and developing treatments for HBV infection. This review summarizes critical findings regarding NTCP's role as a viral receptor for HBV and HDV and discusses important questions that remain unanswered.


Asunto(s)
Virus de la Hepatitis B/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Proteínas Portadoras/metabolismo , Virus de la Hepatitis Delta/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Simportadores/metabolismo
2.
Nature ; 606(7916): 1015-1020, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35545671

RESUMEN

The liver takes up bile salts from blood to generate bile, enabling absorption of lipophilic nutrients and excretion of metabolites and drugs1. Human Na+-taurocholate co-transporting polypeptide (NTCP) is the main bile salt uptake system in liver. NTCP is also the cellular entry receptor of human hepatitis B and D viruses2,3 (HBV/HDV), and has emerged as an important target for antiviral drugs4. However, the molecular mechanisms underlying NTCP transport and viral receptor functions remain incompletely understood. Here we present cryo-electron microscopy structures of human NTCP in complexes with nanobodies, revealing key conformations of its transport cycle. NTCP undergoes a conformational transition opening a wide transmembrane pore that serves as the transport pathway for bile salts, and exposes key determinant residues for HBV/HDV binding to the outside of the cell. A nanobody that stabilizes pore closure and inward-facing states impairs recognition of the HBV/HDV receptor-binding domain preS1, demonstrating binding selectivity of the viruses for open-to-outside over inward-facing conformations of the NTCP transport cycle. These results provide molecular insights into NTCP 'gated-pore' transport and HBV/HDV receptor recognition mechanisms, and are expected to help with development of liver disease therapies targeting NTCP.


Asunto(s)
Ácidos y Sales Biliares , Microscopía por Crioelectrón , Hígado , Transportadores de Anión Orgánico Sodio-Dependiente , Sodio , Simportadores , Bilis/metabolismo , Ácidos y Sales Biliares/metabolismo , Virus de la Hepatitis B/metabolismo , Virus de la Hepatitis Delta/metabolismo , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Transportadores de Anión Orgánico Sodio-Dependiente/química , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Transportadores de Anión Orgánico Sodio-Dependiente/ultraestructura , Conformación Proteica , Receptores Virales/metabolismo , Anticuerpos de Dominio Único , Sodio/metabolismo , Simportadores/química , Simportadores/metabolismo , Simportadores/ultraestructura , Internalización del Virus
3.
PLoS Pathog ; 18(6): e1010633, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35714170

RESUMEN

Hepatitis B virus (HBV) has a highly restricted host range and cell tropism. Other than the human sodium taurocholate cotransporting polypeptide (huNTCP), the HBV entry receptor, host determinants of HBV susceptibility are poorly understood. Woodchucks are naturally infected with woodchuck hepatitis virus (WHV), closely related to HBV, but not with HBV. Here, we investigated the capabilities of woodchuck hepatic and human non-hepatic cell lines to support HBV infection. DNA transfection assays indicated that all cells tested supported both HBV and WHV replication steps post entry, including the viral covalently closed circular DNA (cccDNA) formation, which is essential for establishing and sustaining infection. Ectopic expression of huNTCP rendered one, but not the other, woodchuck hepatic cell line and the non-hepatic human cell line competent to support productive HBV entry, defined here by cccDNA formation during de novo infection. All huNTCP-expressing cell lines tested became susceptible to infection with hepatitis D virus (HDV) that shares the same entry receptor and initial steps of entry with HBV, suggesting that a late entry/trafficking step(s) of HBV infection was defective in one of the two woodchuck cell lines. In addition, the non-susceptible woodchuck hepatic cell line became susceptible to HBV after fusion with human hepatic cells, suggesting the lack of a host cell-dependent factor(s) in these cells. Comparative transcriptomic analysis of the two woodchuck cell lines revealed widespread differences in gene expression in multiple biological processes that may contribute to HBV infection. In conclusion, other than huNTCP, neither human- nor hepatocyte-specific factors are essential for productive HBV entry. Furthermore, a late trafficking step(s) during HBV infection, following the shared entry steps with HDV and before cccDNA formation, is subject to host cell regulation and thus, a host determinant of HBV infection.


Asunto(s)
Virus de la Hepatitis B de la Marmota , Hepatitis B , Animales , ADN Circular/metabolismo , ADN Viral/genética , ADN Viral/metabolismo , Virus de la Hepatitis B/metabolismo , Virus de la Hepatitis Delta/genética , Virus de la Hepatitis Delta/metabolismo , Hepatocitos , Humanos , Marmota , Replicación Viral/genética
4.
J Am Chem Soc ; 145(5): 2830-2839, 2023 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-36706353

RESUMEN

Ribonucleases and small nucleolytic ribozymes are both able to catalyze RNA strand cleavage through 2'-O-transphosphorylation, provoking the question of whether protein and RNA enzymes facilitate mechanisms that pass through the same or distinct transition states. Here, we report the primary and secondary 18O kinetic isotope effects for hepatitis delta virus ribozyme catalysis that reveal a dissociative, metaphosphate-like transition state in stark contrast to the late, associative transition states observed for reactions catalyzed by specific base, Zn2+ ions, or ribonuclease A. This new information provides evidence for a discrete ribozyme active site design that modulates the RNA cleavage pathway to pass through an altered transition state.


Asunto(s)
ARN Catalítico , ARN Catalítico/química , Virus de la Hepatitis Delta/genética , Virus de la Hepatitis Delta/metabolismo , ARN/química , Catálisis , Dominio Catalítico , Conformación de Ácido Nucleico , Cinética
5.
Biochem Biophys Res Commun ; 675: 139-145, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37473528

RESUMEN

Given that the current approved anti-hepatitis B virus (HBV) drugs suppress virus replication and improve hepatitis but cannot eliminate HBV from infected patients, new anti-HBV agents with different mode of action are urgently needed. In this study, we identified a semi-synthetic oxysterol, Oxy185, that can prevent HBV infection in a HepG2-based cell line and primary human hepatocytes. Mechanistically, Oxy185 inhibited the internalization of HBV into cells without affecting virus attachment or replication. We also found that Oxy185 interacted with an HBV entry receptor, sodium taurocholate cotransporting polypeptide (NTCP), and inhibited the oligomerization of NTCP to reduce the efficiency of HBV internalization. Consistent with this mechanism, Oxy185 also inhibited the hepatitis D virus infection, which relies on NTCP-dependent internalization, but not hepatitis A virus infection, and displayed pan-genotypic anti-HBV activity. Following oral administration in mice, Oxy185 showed sustained accumulation in the livers of the mice, along with a favorable liver-to-plasma ratio. Thus, Oxy185 is expected to serve as a useful tool compound in proof-of-principle studies for HBV entry inhibitors with this novel mode of action.


Asunto(s)
Hepatitis B , Simportadores , Humanos , Ratones , Animales , Virus de la Hepatitis B/fisiología , Internalización del Virus , Hepatitis B/metabolismo , Hepatocitos/metabolismo , Células Hep G2 , Virus de la Hepatitis Delta/metabolismo , Simportadores/metabolismo , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo
6.
Biol Chem ; 404(7): 673-690, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37103224

RESUMEN

Na+/taurocholate cotransporting polypeptide (NTCP) is a member of the solute carrier (SLC) family 10 transporters (gene symbol SLC10A1) and is responsible for the sodium-dependent uptake of bile salts across the basolateral membrane of hepatocytes. In addition to its primary transporter function, NTCP is the high-affinity hepatic receptor for hepatitis B (HBV) and hepatitis D (HDV) viruses and, therefore, is a prerequisite for HBV/HDV virus entry into hepatocytes. The inhibition of HBV/HDV binding to NTCP and internalization of the virus/NTCP receptor complex has become a major concept in the development of new antiviral drugs called HBV/HDV entry inhibitors. Hence, NTCP has emerged as a promising target for therapeutic interventions against HBV/HDV infections in the last decade. In this review, recent findings on protein-protein interactions (PPIs) between NTCP and cofactors relevant for entry of the virus/NTCP receptor complex are summarized. In addition, strategies aiming to block PPIs with NTCP to dampen virus tropism and HBV/HDV infection rates are discussed. Finally, this article suggests novel directions for future investigations evaluating the functional contribution of NTCP-mediated PPIs in the development and progression of HBV/HDV infection and subsequent chronic liver disorders.


Asunto(s)
Hepatitis B , Simportadores , Humanos , Antivirales/farmacología , Células Hep G2 , Hepatitis B/tratamiento farmacológico , Hepatitis B/metabolismo , Virus de la Hepatitis B , Virus de la Hepatitis Delta/metabolismo , Hepatocitos/metabolismo , Péptidos , Simportadores/metabolismo , Ácido Taurocólico/metabolismo , Ácido Taurocólico/uso terapéutico , Internalización del Virus
7.
J Hepatol ; 74(3): 686-699, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33276031

RESUMEN

Chronic hepatitis D (CHD), a global health problem, manifests as the most severe form of viral hepatitis. The causative agent, HDV, is the smallest known human virus; it replicates its circular single-stranded RNA genome in the nucleus of hepatocytes. HDV requires HBV-encoded envelope proteins for dissemination and de novo cell entry. However, HDV can also spread through cell division. Following entry into hepatocytes, replicative intermediates of HDV RNA are sensed by the pattern recognition receptor MDA5 (melanoma differentiation antigen 5) resulting in interferon (IFN)-ß/λ induction. This IFN response strongly suppresses cell division-mediated spread of HDV genomes, however, it only marginally affects HDV RNA replication in already infected, resting hepatocytes. Monotherapy with IFN-α/λ shows efficacy but rarely results in HDV clearance. Recent molecular insights into key determinants of HDV persistence and the accelerated development of specifically acting antivirals that interfere with the replication cycle have revealed promising new therapeutic perspectives. In this review, we briefly summarise our knowledge on replication/persistence of HDV, the newly discovered HDV-like agents, and the interplay of HDV with the IFN response and its consequences for persistence. Finally, we discuss the possible role of IFNs in combination with upcoming therapies aimed at HDV cure.


Asunto(s)
Antivirales/uso terapéutico , Hepatitis D Crónica/tratamiento farmacológico , Hepatitis D Crónica/inmunología , Virus de la Hepatitis Delta/metabolismo , Inmunidad Innata , Interferón-alfa/metabolismo , Animales , Antivirales/farmacología , División Celular/efectos de los fármacos , Hepatitis B/complicaciones , Hepatitis B/virología , Virus de la Hepatitis B/metabolismo , Hepatitis D Crónica/complicaciones , Hepatitis D Crónica/virología , Virus de la Hepatitis Delta/genética , Hepatocitos/metabolismo , Hepatocitos/virología , Humanos , Interferón beta/metabolismo , Resultado del Tratamiento , Replicación Viral/efectos de los fármacos
8.
J Virol ; 94(4)2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-31748400

RESUMEN

A substantial number of viruses have been demonstrated to subvert autophagy to promote their own replication. Recent publications have reported the proviral effect of autophagy induction on hepatitis B virus (HBV) replication. Hepatitis delta virus (HDV) is a defective virus and an occasional obligate satellite of HBV. However, no previous work has studied the relationship between autophagy and HDV. In this article, we analyze the impact of HBV and HDV replication on autophagy as well as the involvement of the autophagy machinery in the HDV life cycle when produced alone and in combination with HBV. We prove that HBxAg and HBsAg can induce early steps of autophagy but ultimately block flux. It is worth noting that the two isoforms of the HDV protein, the small HDAg (S-HDAg) and large HDAg (L-HDAg) isoforms, can also efficiently promote autophagosome accumulation and disturb autophagic flux. Using CRISPR-Cas9 technology to generate specific knockouts, we demonstrate that the autophagy machinery, specifically the proteins implicated in the elongation step (ATG7, ATG5, and LC3), is important for the release of HBV without affecting the level of intracellular HBV genomes. Surprisingly, the knockout of ATG5 and ATG7 decreased the intracellular HDV RNA level in both Huh7 and HepG2.2.15 cells without an additional effect on HDV secretion. Therefore, we conclude that HBV and HDV have evolved to utilize the autophagy machinery so as to assist at different steps of their life cycle.IMPORTANCE Hepatitis delta virus is a defective RNA virus that requires hepatitis B virus envelope proteins (HBsAg) to fulfill its life cycle. Thus, HDV can only infect individuals at the same time as HBV (coinfection) or superinfect individuals who are already chronic carriers of HBV. The presence of HDV in the liver accelerates the progression of infection to fibrosis and to hepatic cancer. Since current treatments against HBV are ineffective against HDV, it is of paramount importance to study the interaction between HBV, HDV, and host factors. This will help unravel new targets whereby a therapy that is capable of simultaneously impeding both viruses could be developed. In this research paper, we evidence that the autophagy machinery promotes the replication of HBV and HDV at different steps of their life cycle. Notwithstanding their contribution to HBV release, autophagy proteins seem to assist HDV intracellular replication but not its secretion.


Asunto(s)
Autofagia/genética , Virus de la Hepatitis Delta/metabolismo , Replicación Viral/fisiología , Línea Celular , Coinfección/virología , Células HEK293 , Células Hep G2 , Hepatitis B/virología , Antígenos de Superficie de la Hepatitis B/genética , Virus de la Hepatitis B/genética , Hepatitis D/virología , Virus de la Hepatitis Delta/genética , Antígenos de Hepatitis delta/metabolismo , Humanos , Hígado/metabolismo , ARN Viral/genética
9.
J Org Chem ; 86(19): 13231-13244, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34533968

RESUMEN

Oligoribonucleotides containing a photocaged 2'-amino-5'-S-phophorothiolate linkage have potential applications as therapeutic agents and biological probes to investigate the RNA structure and function. We envisioned that oligoribonucleotides containing a 2'-amino-5'-S-phosphorothiolate linkage could provide an approach to identify the general base within catalytic RNAs by chemogenetic suppression. To enable preliminary tests of this idea, we developed synthetic approaches to a dinucleotide, trinucleotide, and oligoribonucleotide containing a photocaged 2'-amino-5'-S-phosphorothiolate linkage. We incorporated the photocaged 2'-amino-5'-S-phosphorothiolate linkage into an oligoribonucleotide substrate for the hepatitis delta virus (HDV) ribozyme and investigated the pH dependence of its cleavage following UV irradiation both in the presence and absence of the ribozyme. The substrate exhibited a pH-rate profile characteristic of the modified linkage but reacted slower when bound to the ribozyme. Cleavage inhibition by the HDV ribozyme could reflect a non-productive ground-state interaction with the modified substrate's nucleophilic 2'-NH2 or a poor fit of the modified transition state at the ribozyme's active site.


Asunto(s)
Oligorribonucleótidos , ARN Catalítico , Dominio Catalítico , Virus de la Hepatitis Delta/genética , Virus de la Hepatitis Delta/metabolismo , ARN , ARN Catalítico/metabolismo
10.
Gut ; 68(1): 150-157, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29217749

RESUMEN

OBJECTIVE: Hepatitis delta virus (HDV) was shown to persist for weeks in the absence of HBV and for months after liver transplantation, demonstrating the ability of HDV to persevere in quiescent hepatocytes. The aim of the study was to evaluate the impact of cell proliferation on HDV persistence in vitro and in vivo. DESIGN: Genetically labelled human sodium taurocholate cotransporting polypeptide (hNTCP)-transduced human hepatoma(HepG2) cells were infected with HBV/HDV and passaged every 7 days for 100 days in the presence of the entry inhibitor Myrcludex-B. In vivo, cell proliferation was triggered by transplanting primary human hepatocytes (PHHs) isolated from HBV/HDV-infected humanised mice into naïve recipients. Virological parameters were measured by quantitative real time polymerase chain reaction (qRT-PCR). Hepatitis delta antigen (HDAg), hepatitis B core antigen (HBcAg) and cell proliferation were determined by immunofluorescence. RESULTS: Despite 15 in vitro cell passages and block of viral spreading by Myrcludex-B, clonal cell expansion permitted amplification of HDV infection. In vivo, expansion of PHHs isolated from HBV/HDV-infected humanised mice was confirmed 3 days, 2, 4 and 8 weeks after transplantation. While HBV markers rapidly dropped in proliferating PHHs, HDAg-positive hepatocytes were observed among dividing cells at all time points. Notably, HDAg-positive cells appeared in clusters, indicating that HDV was transmitted to daughter cells during liver regeneration even in the absence of de novo infection. CONCLUSION: This study demonstrates that HDV persists during liver regeneration by transmitting HDV RNA to dividing cells even in the absence of HBV coinfection. The strong persistence capacities of HDV may also explain why HDV clearance is difficult to achieve in HBV/HDV chronically infected patients.


Asunto(s)
Coinfección/virología , Hepatitis B/virología , Hepatitis D/virología , Virus de la Hepatitis Delta/metabolismo , Regeneración Hepática , Animales , División Celular , Línea Celular , Proliferación Celular , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , ARN Viral/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
11.
Biochem Cell Biol ; 97(2): 130-139, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30153423

RESUMEN

Liver coinfection by hepatitis B virus (HBV) and hepatitis D virus (HDV) can result in a severe form of hepatocellular carcinoma with poor prognosis. Coinfection with HDV and HBV causes more deleterious effects than infection with HBV alone. Clinical research has shown that glutathione S-transferase P1 (GSTP1), a tumor suppressor gene, is typically downregulated in liver samples from hepatitis-infected patients. In the present study, our data indicated that small HDV antigen (s-HDAg) could specifically bind to GSTP1 mRNA and significantly downregulate GSTP1 protein expression. For the human fetal hepatocyte cell line L-02, cells transfected with s-HDAg, along with decreased GSTP1 expression, there was a significant accumulation of reactive oxygen species (ROS) and increased apoptotic ratios. Restoring GSTP1 expression through silencing s-HDAg via RNAi or overexpressing exogenous GSTP1 could largely recover the abnormal cell status. Our results revealed a novel potential mechanism of HDV-induced liver injury and hepatocarcinogenesis: s-HDAg can inhibit GSTP1 expression by directly binding to GSTP1 mRNA, which leads to accumulation of cellular ROS, resulting in high cellular apoptotic ratios and increased selective pressure for malignant transformation. To our knowledge, this is the first study to examine s-HDAg-specific pathogenic mechanisms through potential protein-RNA interactions.


Asunto(s)
Transformación Celular Viral , Regulación hacia Abajo , Regulación Enzimológica de la Expresión Génica , Gutatión-S-Transferasa pi/biosíntesis , Virus de la Hepatitis Delta/metabolismo , Antígenos de Hepatitis delta/metabolismo , Neoplasias Hepáticas/metabolismo , Hígado/metabolismo , ARN Mensajero/metabolismo , Línea Celular , Gutatión-S-Transferasa pi/genética , Virus de la Hepatitis Delta/genética , Antígenos de Hepatitis delta/genética , Humanos , Hígado/lesiones , Hígado/patología , Hígado/virología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , ARN Mensajero/genética
12.
Biochemistry ; 57(25): 3465-3472, 2018 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-29733591

RESUMEN

Steric constraints imposed by the active sites of protein and RNA enzymes pose major challenges to the investigation of structure-function relationships within these systems. As a strategy to circumvent such constraints in the HDV ribozyme, we have synthesized phosphoramidites from propanediol derivatives and incorporated them at the 5'-termini of RNA and DNA oligonucleotides to generate a series of novel substrates with nucleophiles perturbed electronically through geminal fluorination. In nonenzymatic, hydroxide-catalyzed intramolecular transphosphorylation of the DNA substrates, pH-rate profiles revealed that fluorine substitution reduces the maximal rate and the kinetic p Ka, consistent with the expected electron-withdrawing effect. In HDV ribozyme reactions, we observed that the RNA substrates undergo transphosphorylation relatively efficiently, suggesting that the conformational constraints imposed by a ribofuranose ring are not strictly required for ribozyme catalysis. In contrast to the nonenzymatic reactions, however, substrate fluorination modestly increases the ribozyme reaction rate, consistent with a mechanism in which (1) the 2'-hydroxyl nucleophile exists predominantly in its neutral, protonated form in the ground state and (2) the 2'-hydroxyl bears some negative charge in the rate-determining step, consistent with a transition state in which the extent of 2'-OH deprotonation exceeds the extent of P-O bond formation.


Asunto(s)
Hepatitis D/virología , Virus de la Hepatitis Delta/enzimología , ARN Catalítico/metabolismo , ARN Viral/metabolismo , ADN/química , ADN/metabolismo , Virus de la Hepatitis Delta/química , Virus de la Hepatitis Delta/metabolismo , Humanos , Conformación de Ácido Nucleico , Oligonucleótidos/química , Oligonucleótidos/metabolismo , Compuestos Organofosforados/química , Compuestos Organofosforados/metabolismo , Protones , ARN Catalítico/química , ARN Viral/química , Especificidad por Sustrato
13.
ScientificWorldJournal ; 2018: 9312650, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30356409

RESUMEN

BACKGROUND: Hepatitis D virus (HDV) infection has been considered a serious neglected pandemic, particularly in developing countries. The virus causes a more severe disease than mono infection with hepatitis B virus (HBV). The epidemiology of HDV is not well documented in North Africa, which is known to be endemic for HBV. In this study, we explored the prevalence of HDV infection and also attempted to identify factors associated with hepatitis D positive status among chronic hepatitis B patients in North Africa. METHODS: The electronic databases PubMed, Embase, Scopus, Science Direct, Web of Science, and Google Scholar were comprehensively searched for all papers published between January 1, 1998, and December 31, 2017, using appropriate strategies containing all related keywords, including North Africa, names of countries in the region, and all permutations of hepatitis D virus. The estimated prevalence of HDV in North Africa was calculated as an average of the pooled infection prevalence in each country weighted by the ratio of the country's hepatitis D virus population to the study's sample size in the survey data analysis. FINDINGS: A total of 312 studies were identified and 32 were included in this study, with a total sample of 4907 individuals screened for HDV. There was considerable variability in the prevalence estimates of HDV within the countries of the region. The overall prevalence of HDV in the general population of North Africa was 5·01% (95% CI: 1·25-8·27) and in liver disease patients it was 20.7% (95% CI:9.87-44.53). Genotype-1 was the most prominent genotype reported in five published studies. Ten studies reported on HDV RNA in participants who were seropositive for HDV, and four studies highlighted the impact of demographic factors (sex and age). No study showed the impact of risk factors on the prevalence of HDV in North Africa. INTERPRETATION: This review provides a comprehensive assessment of the burden of HDV in Northern Africa. There were significant differences in seroprevalence, study population, and diagnostic testing between the countries in the region. The results presented here will alert health professionals to implement clear policies based on evidence to diminish the burden of HDV infection. Such measures may include but are not restricted to improving the laboratory diagnostic tests and initiating patient data registries and blood screening. Further epidemiological and research studies are needed to explore the risk factors, coinfections, and approaches to increase testing for HDV, particularly in high-risk subpopulations, such as intravenous drug users and immigrants, and to define the consequences of HDV infection in North Africa.


Asunto(s)
Costo de Enfermedad , Hepatitis D/epidemiología , Virus de la Hepatitis Delta/aislamiento & purificación , África del Norte/epidemiología , Bases de Datos Factuales/tendencias , Emigración e Inmigración/tendencias , Hepatitis D/sangre , Hepatitis D/diagnóstico , Virus de la Hepatitis Delta/metabolismo , Humanos , Abuso de Sustancias por Vía Intravenosa/sangre , Abuso de Sustancias por Vía Intravenosa/diagnóstico , Abuso de Sustancias por Vía Intravenosa/epidemiología
14.
Artículo en Ruso | MEDLINE | ID: mdl-30695348

RESUMEN

AIM: Evaluate significance of covalently closed circular DNA of hepatitis B virus as a marker for detection of occult viral hepatitis B in Uzbekistan population with hepatitis of various genesis. MATERIALS AND METHODS: Blood plasma and liver biopsy from 39 patients with different severity levels of liver fibrosis and cirrhosis served as study material. HBV covalently closed circular DNA detection was carried out according to Pollicino T et al. (2004). RESULTS: Covalently closed circu- lar DNA of hepatitis B virus was detected in 82% of samples, including in 54.5% of patients with chronic viral hepatitis C (CVHC) and in 100% of patients with hepatitis of unknown etiology. Quantitative evaluation of content of covalently closed circular DNA of hepatitis B virus in liver tissue in patients with CVHB has shown an average of 2.5 copies of HBV genome as ccc DNA per cell, in patients with CVHB + D an average of 0.7 copies/cell, in patients with co-infection by HCV and HBV - 0.5 copies/cell, in patients with CVHC an average of 0.12 copies/cell, and in patients with cryptogenic hepatitis - 0.2 copies/cell. CONCLUSION: Detection of HBV DNA is a complex problem for effective laboratory diagnostics of hepatitis. Detection of HBV ccc DNA as a marker of occult hepatitis B in patients with CVHC and patients with hepatitis of unclear etiol- ogy is an. important factor for diagnostics, selection of adequate therapy, prognosis of disease outcome and prevention of development of severe liver diseases.


Asunto(s)
ADN Circular/metabolismo , ADN Viral/metabolismo , Hepacivirus/metabolismo , Virus de la Hepatitis B/metabolismo , Hepatitis B/metabolismo , Hepatitis C Crónica/metabolismo , Hepatitis D/metabolismo , Virus de la Hepatitis Delta/metabolismo , Femenino , Hepatitis B/epidemiología , Hepatitis C Crónica/epidemiología , Hepatitis D/epidemiología , Humanos , Masculino , Prevalencia , Uzbekistán/epidemiología
15.
J Am Chem Soc ; 137(28): 8973-82, 2015 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-26125657

RESUMEN

Endonucleolytic ribozymes constitute a class of non-coding RNAs that catalyze single-strand RNA scission. With crystal structures available for all of the known ribozymes, a major challenge involves relating functional data to the physically observed RNA architecture. In the case of the hepatitis delta virus (HDV) ribozyme, there are three high-resolution crystal structures, the product state of the reaction and two precursor variants, with distinct mechanistic implications. Here, we develop new strategies to probe the structure and catalytic mechanism of a ribozyme. First, we use double-mutant cycles to distinguish differences in functional group proximity implicated by the crystal structures. Second, we use a corrected form of the Brønsted equation to assess the functional significance of general acid catalysis in the system. Our results delineate the functional relevance of atomic interactions inferred from structure, and suggest that the HDV ribozyme transition state resembles the cleavage product in the degree of proton transfer to the leaving group.


Asunto(s)
Hepatitis D/virología , Virus de la Hepatitis Delta/enzimología , ARN Catalítico/metabolismo , ARN Viral/metabolismo , Secuencia de Bases , Dominio Catalítico , Virus de la Hepatitis Delta/química , Virus de la Hepatitis Delta/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Protones , ARN Catalítico/química , ARN Viral/química
16.
J Virol ; 88(16): 9049-59, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24899172

RESUMEN

UNLABELLED: The envelope proteins of hepatitis B virus (HBV) bear an N-linked glycosylation site at N146 within the immunodominant a-determinant in the antigenic loop (AGL) region. This glycosylation site is never fully functional, leading to a nearly 1/1 ratio of glycosylated/nonglycosylated isoforms in the viral envelope. Here we investigated the requirement for a precise positioning of N-linked glycan at amino acid 146 and the functions associated with the glycosylated and nonglycosylated isoforms. We observed that the removal of the N146 glycosylation site by mutagenesis was permissive to envelope protein synthesis and stability and to secretion of subviral particles (SVPs) and hepatitis delta virus (HDV) virions, but it was detrimental to HBV virion production. Several positions in the AGL could substitute for position 146 as the glycosylation acceptor site. At position 146, neither a glycan chain nor asparagine was absolutely required for infectivity, but there was a preference for a polar residue. Envelope proteins bearing 5 AGL glycosylation sites became hyperglycosylated, leading to an increased capacity for SVP secretion at the expense of HBV and HDV virion secretion. Infectivity-compatible N-glycosylation sites could be inserted at 3 positions (positions 115, 129, and 136), but when all three positions were glycosylated, the hyperglycosylated mutant was substantially attenuated at viral entry, while it acquired resistance to neutralizing antibodies. Taken together, these findings suggest that the nonglycosylated N146 is essential for infectivity, while the glycosylated form, in addition to its importance for HBV virion secretion, is instrumental in shielding the a-determinant from neutralizing antibodies. IMPORTANCE: At the surface of HBV particles, the immunodominant a-determinant is the main target of neutralizing antibodies and an essential determinant of infectivity. It contains an N-glycosylation site at position 146, which is functional on only half of the envelope proteins. Our data suggest that the coexistence of nonglycosylated and glycosylated N146 at the surface of HBV reflects the dual function of this determinant in infectivity and immune escape. Hence, a modification of the HBV glycosylation pattern affects not only virion assembly and infectivity but also immune escape.


Asunto(s)
Virus de la Hepatitis B/genética , Virus de la Hepatitis B/inmunología , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Virión/metabolismo , Secuencia de Aminoácidos , Anticuerpos Neutralizantes/genética , Anticuerpos Neutralizantes/inmunología , Línea Celular , Línea Celular Tumoral , Glicosilación , Hepatitis B/inmunología , Hepatitis B/metabolismo , Hepatitis B/virología , Virus de la Hepatitis B/metabolismo , Hepatitis D/inmunología , Hepatitis D/metabolismo , Hepatitis D/virología , Virus de la Hepatitis Delta/genética , Virus de la Hepatitis Delta/inmunología , Virus de la Hepatitis Delta/metabolismo , Humanos , Datos de Secuencia Molecular , Mutagénesis/genética , Mutagénesis/inmunología , Proteínas del Envoltorio Viral/inmunología , Virión/genética , Virión/inmunología , Ensamble de Virus/genética , Ensamble de Virus/inmunología , Internalización del Virus
17.
Anal Biochem ; 483: 12-20, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25937290

RESUMEN

Biological catalysis involves interactions distant from the site of chemistry that can position the substrate for reaction. Catalysis of RNA 2'-O-transphosphorylation by the hepatitis delta virus (HDV) ribozyme is sensitive to the identity of the N(-1) nucleotide flanking the reactive phosphoryl group. However, the interactions that affect the conformation of this position, and in turn the 2'O nucleophile, are unclear. Here, we describe the application of multiple substrate internal competition kinetic analyses to understand how the N(-1) nucleobase contributes to HDV catalysis and test the utility of this approach for RNA structure-function studies. Internal competition reactions containing all four substrate sequence variants at the N(-1) position in reactions using ribozyme active site mutations at A77 and A78 were used to test a proposed base-pairing interaction. Mutants A78U, A78G, and A79G retain significant catalytic activity but do not alter the specificity for the N(-1) nucleobase. Effects of nucleobase analog substitutions at N(-1) indicate that U is preferred due to the ability to donate an H-bond in the Watson-Crick face and avoid minor groove steric clash. The results provide information essential for evaluating models of the HDV active site and illustrate multiple substrate kinetic analyses as a practical approach for characterizing structure-function relationships in RNA reactions.


Asunto(s)
Virus de la Hepatitis Delta/metabolismo , ARN Catalítico/metabolismo , Emparejamiento Base , Secuencia de Bases , Electroforesis en Gel de Poliacrilamida , Genoma Viral , Enlace de Hidrógeno , Cinética , Datos de Secuencia Molecular , Mutagénesis , Conformación de Ácido Nucleico , ARN Catalítico/química , ARN Catalítico/genética , Especificidad por Sustrato , Uridina
18.
Phys Chem Chem Phys ; 17(8): 5887-900, 2015 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-25631765

RESUMEN

The hepatitis delta virus (HDV) is the only known human pathogen whose genome contains a catalytic RNA motif (ribozyme). The overall architecture of the HDV ribozyme is that of a double-nested pseudoknot, with two GU pairs flanking the active site. Although extensive studies have shown that mutation of either wobble results in decreased catalytic activity, little work has focused on linking these mutations to specific structural effects on catalytic fitness. Here we use molecular dynamics simulations based on an activated structure to probe the active site dynamics as a result of wobble pair mutations. In both wild-type and mutant ribozymes, the in-line fitness of the active site (as a measure of catalytic proficiency) strongly depends on the presence of a C75(N3H3+)N1(O5') hydrogen bond, which positions C75 as the general acid for the reaction. Our mutational analyses show that each GU wobble supports catalytically fit conformations in distinct ways; the reverse G25U20 wobble promotes high in-line fitness, high occupancy of the C75(N3H3+)G1(O5') general-acid hydrogen bond and stabilization of the G1U37 wobble, while the G1U37 wobble acts more locally by stabilizing high in-line fitness and the C75(N3H3+)G1(O5') hydrogen bond. We also find that stable type I A-minor and P1.1 hydrogen bonding above and below the active site, respectively, prevent local structural disorder from spreading and disrupting global conformation. Taken together, our results define specific, often redundant architectural roles for several structural motifs of the HDV ribozyme active site, expanding the known roles of these motifs within all HDV-like ribozymes and other structured RNAs.


Asunto(s)
Virus de la Hepatitis Delta/metabolismo , ARN Catalítico/química , Emparejamiento Base , Catálisis , Dominio Catalítico , Virus de la Hepatitis Delta/genética , Humanos , Enlace de Hidrógeno , Magnesio/química , Simulación de Dinámica Molecular , Mutación , Conformación de Ácido Nucleico , ARN Catalítico/genética , ARN Catalítico/metabolismo , Sodio/química
19.
RNA ; 18(12): 2157-65, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23105000

RESUMEN

Protein Kinase R (PKR), the double-stranded RNA (dsRNA)-activated protein kinase, plays important roles in innate immunity. Previous studies have shown that PKR is activated by long stretches of dsRNA, RNA pseudoknots, and certain single-stranded RNAs; however, regulation of PKR by RNAs with globular tertiary structure has not been reported. In this study, the HDV ribozyme is used as a model of a mostly globular RNA. In addition to a catalytic core, the ribozyme contains a peripheral 13-bp pairing region (P4), which, upon shortening, affects neither the catalytic activity of the ribozyme nor its ability to crystallize. We report that the HDV ribozyme sequence alone can activate PKR. To elucidate the RNA structural basis for this, we prepared a number of HDV variants, including those with shortened or lengthened P4 pairing regions, with the anticipation that lengthening the P4 extension would yield a more potent activator since it would offer more base pairs of dsRNA. Surprisingly, the variant with a shortened P4 was the most potent activator. Through native gel mobility and enzymatic structure mapping experiments we implicate misfolded HDV ribozyme dimers as the PKR-activating species, and show that the shortened P4 leads to enhanced occupancy of the RNA dimer. These observations have implications for how RNA misfolding relates to innate immune response and human disease.


Asunto(s)
Virus de la Hepatitis Delta/metabolismo , Pliegue del ARN/fisiología , ARN Catalítico/química , ARN Catalítico/metabolismo , eIF-2 Quinasa/metabolismo , Secuencia de Bases , Dimerización , Activación Enzimática , Virus de la Hepatitis Delta/genética , Humanos , Técnicas In Vitro , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , ARN Catalítico/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , eIF-2 Quinasa/genética
20.
J Virol ; 87(14): 7977-91, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23678176

RESUMEN

Human hepatitis B virus (HBV) and its satellite virus, hepatitis D virus (HDV), primarily infect humans, chimpanzees, or tree shrews (Tupaia belangeri). Viral infections in other species are known to be mainly restricted at the entry level since viral replication can be achieved in the cells by transfection of the viral genome. Sodium taurocholate cotransporting polypeptide (NTCP) is a functional receptor for HBV and HDV, and amino acids 157 to 165 of NTCP are critical for viral entry and likely limit viral infection of macaques. However, the molecular determinants for viral entry restriction in mouse NTCP (mNTCP) remain unclear. In this study, mNTCP was found to be unable to support either HBV or HDV infection, although it can bind to pre-S1 of HBV L protein and is functional in transporting substrate taurocholate; comprehensive swapping and point mutations of human NTCP (hNTCP) and mNTCP revealed molecular determinants restricting mNTCP for viral entry of HBV and HDV. Remarkably, when mNTCP residues 84 to 87 were substituted by human counterparts, mNTCP can effectively support viral infections. In addition, a number of cell lines, regardless of their species or tissue origin, supported HDV infection when transfected with hNTCP or mNTCP with residues 84 to 87 replaced by human counterparts, highlighting the central role of NTCP for viral infections mediated by HBV envelope proteins. These studies advance our understanding of NTCP-mediated viral entry of HBV and HDV and have important implications for developing the mouse model for their infections.


Asunto(s)
Virus de la Hepatitis B/metabolismo , Virus de la Hepatitis Delta/metabolismo , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Simportadores/metabolismo , Internalización del Virus , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Ensayo de Inmunoadsorción Enzimática , Fluoresceína-5-Isotiocianato , Antígenos de Superficie de la Hepatitis B/metabolismo , Humanos , Ratones , Microscopía Fluorescente , Mutación/genética , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Precursores de Proteínas/metabolismo , Simportadores/genética , Ácido Taurocólico/metabolismo , Tritio , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA