Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Virol ; 97(1): e0171722, 2023 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-36475867

RESUMO

Hepatitis B virus (HBV) infection remains a public health problem worldwide. Persistent HBV infection relies on active transcription of the covalently closed circular DNA (cccDNA) in hepatocytes, which is less understood at the single-cell level. In this study, we isolated primary human hepatocytes from liver-humanized FRG mice infected with HBV and examined cccDNA transcripts in single cells based on 5' end sequencing. Our 5' transcriptome sequencing (RNA-seq) analysis unambiguously assigns different viral transcripts with overlapping 3' sequences and quantitatively measures viral transcripts for structural genes (3.5 kb, 2.4 kb, and 2.1 kb) and the nonstructural X gene (0.7 kb and related) in single cells. We found that an infected cell either can generate all viral transcripts, signifying active transcription, or presents only transcripts from the X gene and its associated enhancer I domain and no structural gene transcripts. Results from cell infection assays with recombinant HBV show that nonproductive transcription of cccDNA can be activated by incoming virus through superinfection. Moreover, upon HBV infection, cccDNA apparently can be transcribed in the absence of HBx and produces HBx, needed for productive transcription of other viral genes. These results shed new light on cccDNA transcription at the single-cell level and provide insights useful for improving the treatment strategy against chronic HBV infection. IMPORTANCE Hepatitis B virus (HBV) infection can be effectively suppressed but rarely cured by available drugs. Chronic HBV infection is based on persistence of covalently closed circular DNA (cccDNA) and continuous infection and reinfection with HBV in the liver. Understanding transcriptional regulation of cccDNA will help to achieve permanent transcriptional silencing, i.e., functional cure of HBV. In our study, we found that an infected cell either can generate all viral transcripts, signifying active transcription, or presents only transcripts from the X gene and its associated enhancer I domain and no structural gene transcripts. The nonproductive transcription of cccDNA can be activated by incoming virus through superinfection. Upon an infection, cccDNA apparently can be transcribed in the absence of HBx to produce HBx, necessary for subsequent transcription of other HBV genes. Our studies shed new light on the mechanism of HBV infection and may have implications for a functional cure regimen for HBV.


Assuntos
DNA Circular , Hepatite B Crônica , Superinfecção , Animais , Humanos , Camundongos , DNA Circular/genética , DNA Viral/genética , Vírus da Hepatite B/genética , Hepatite B Crônica/genética , Replicação Viral/genética , Hepatócitos , Proteínas Virais Reguladoras e Acessórias/genética
2.
Nat Commun ; 12(1): 2186, 2021 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-33846305

RESUMO

To gain mechanistic insights into the functions and developmental dynamics of tumor-infiltrated immune cells, especially B-lymphocytes, here we combine single-cell RNA-sequencing and antigen receptor lineage analysis to characterize a large number of triple-negative breast cancer infiltrated immune cells and report a comprehensive atlas of tumor-infiltrated B-lymphocytes. The single-cell transcriptional profiles reveal significant heterogeneity in tumor-infiltrated B-cell subgroups. The single-cell antigen receptor analyses demonstrate that compared with those in peripheral blood, tumor-infiltrated B-cells have more mature and memory B-cell characteristics, higher clonality, more class switching recombination and somatic hypermutations. Combined analyses suggest local differentiation of infiltrated memory B-cells within breast tumors. The B-cell signatures based on the single-cell RNA-sequencing results are significantly associated with improved survival in breast tumor patients. Functional analyses of tumor-infiltrated B-cell populations suggest that mechanistically, B-cell subgroups may contribute to immunosurveillance through various pathways. Further dissection of tumor-infiltrated B-cell populations will provide valuable clues for tumor immunotherapy.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Perfilação da Expressão Gênica , Linfócitos do Interstício Tumoral/imunologia , Receptores de Antígenos/metabolismo , Análise de Sequência de RNA , Análise de Célula Única , Idoso , Neoplasias da Mama/sangue , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Cadeias Pesadas de Imunoglobulinas/genética , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Transcriptoma/genética
3.
J Biol Chem ; 294(31): 11853-11862, 2019 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-31201272

RESUMO

Sodium taurocholate cotransporting polypeptide (NTCP, encoded by Slc10a1/SLC10A1) deficiency can result in hypercholanemia but no obvious symptoms in both mice and humans. However, the consequence of and response to long-term hypercholanemia caused by NTCP deficiency remain largely unexplored. Here, we analyzed lifelong dynamics of serum total bile acid (TBA) levels in Slc10a1-/- mice, and we also assessed changes of TBA levels in 33 young individuals with SLC10A1 loss-of-function variant p.Ser267Phe. We found that overall serum TBA levels tended to decrease gradually with age in both Slc10a1-/- mice and p.Ser267Phe individuals. Liver mRNA profiling revealed notable transcription alterations in hypercholanemic Slc10a1-/- mice, including inhibition of bile acid (BA) synthesis, enhancement of BA detoxification, and altered BA transport. Members of the sulfotransferase (SULT) family showed the most dramatic increases in livers of hypercholanemic Slc10a1-/- mice, and one of their BA sulfates, taurolithocholic acid 3-sulfate, significantly increased. Importantly, consistent with the mouse studies, comprehensive profiling of 58 BA species in sera of p.Ser267Phe individuals revealed a markedly increased level of BA sulfates. Together, our findings indicate that the enhanced BA sulfation is a major mechanism for BA detoxification and elimination in both mice and humans with Slc10a1/SLC10A1 deficiency.


Assuntos
Ácidos e Sais Biliares/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Simportadores/genética , Ácido Taurolitocólico/análogos & derivados , Animais , Ácidos e Sais Biliares/sangue , Cromatografia Líquida de Alta Pressão , Feminino , Homozigoto , Humanos , Hipercolesterolemia/patologia , Hipercolesterolemia/veterinária , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Transportadores de Ânions Orgânicos Dependentes de Sódio/deficiência , Simportadores/deficiência , Espectrometria de Massas em Tandem , Ácido Taurolitocólico/sangue , Ácido Taurolitocólico/metabolismo , Ácido Taurolitocólico/urina
4.
Virology ; 522: 168-176, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30032030

RESUMO

Sodium taurocholate cotransporting polypeptide (NTCP) is a functional receptor for human hepatitis B virus (HBV) and its satellite virus Hepatitis D virus (HDV). Physiologically, NTCP is responsible for the majority of sodium-dependent bile acids uptake by hepatocytes. The p.Ser267Phe (S267F) variant of NTCP is a single nucleotide polymorphism (SNP) previously found to cause substantial loss of ability to support HBV and HDV infection and its taurocholic acid uptake function in vitro. Intriguingly, ten individuals were identified as S267F homozygotes in population studies of chronic hepatitis B (CHB) patients. In this study, we identified new HBV isolates from one homozygous S267F mutation carrier and confirmed new isolates also use wildtype-NTCP as a cellular receptor. Furthermore, we demonstrated S267F variant of NTCP, though inefficient, is still a functional receptor for HBV entry. This study advances our understanding of NTCP-mediated HBV infection.


Assuntos
Vírus da Hepatite B/crescimento & desenvolvimento , Hepatite B Crônica/virologia , Interações Hospedeiro-Patógeno , Proteínas Mutantes/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Receptores Virais/metabolismo , Simportadores/metabolismo , Linhagem Celular , Hepatite B Crônica/genética , Hepatócitos/virologia , Homozigoto , Humanos , Proteínas Mutantes/genética , Mutação de Sentido Incorreto , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Receptores Virais/genética , Simportadores/genética , Internalização do Vírus
5.
J Virol ; 92(8)2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29437960

RESUMO

Multiple steps of the life cycle of hepatitis B virus (HBV) are known to be coupled to hepatic metabolism. However, the details of involvement of the hepatic metabolic milieu in HBV infection remain incompletely understood. Hepatic lipid metabolism is controlled by a complicated transcription factor network centered on retinoid X receptor alpha (RXRα). Here, we report that RXRα negatively regulates HBV infection at an early stage in cell cultures. The RXR-specific agonist bexarotene inhibits HBV in HepG2 cells expressing the sodium taurocholate cotransporting polypeptide (NTCP) (HepG2-NTCP), HepaRG cells, and primary Tupaia hepatocytes (PTHs); reducing RXRα expression significantly enhanced HBV infection in the cells. Transcriptome sequencing (RNA-seq) analysis of HepG2-NTCP cells with a disrupted RXRα gene revealed that reduced gene expression in arachidonic acid (AA)/eicosanoid biosynthesis pathways, including the AA synthases phospholipase A2 group IIA (PLA2G2A), is associated with increased HBV infection. Moreover, exogenous treatment of AA inhibits HBV infection in HepG2-NTCP cells. These data demonstrate that RXRα is an important cellular factor in modulating HBV infection and implicate the participation of AA/eicosanoid biosynthesis pathways in the regulation of HBV infection.IMPORTANCE Understanding how HBV infection is connected with hepatic lipid metabolism may provide new insights into virus infection and its pathogenesis. By a series of genetic studies in combination with transcriptome analysis and pharmacological assays, we here investigated the role of cellular retinoid X receptor alpha (RXRα), a crucial transcription factor for controlling hepatic lipid metabolism, in de novo HBV infection in cell cultures. We found that silencing of RXRα resulted in elevated HBV covalently closed circular DNA (cccDNA) formation and viral antigen production, while activation of RXRα reduced HBV infection efficiency. Our results also showed that silencing phospholipase A2 group IIA (PLA2G2A), a key enzyme of arachidonic acid (AA) synthases, enhanced HBV infection efficiency in HepG2-NTCP cells and that exogenous AA treatment reduced de novo HBV infection in the cells. These findings unveil RXRα as an important cellular factor in modulating HBV infection and may point to a new strategy for host-targeted therapies against HBV.


Assuntos
Regulação da Expressão Gênica , Vírus da Hepatite B/metabolismo , Hepatite B/metabolismo , Receptor X Retinoide alfa/biossíntese , Animais , Células Hep G2 , Hepatite B/genética , Humanos , Tupaia
6.
Virology ; 505: 1-11, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28213271

RESUMO

Sodium taurocholate cotransporting polypeptide (NTCP) is the functional receptor for human hepatitis B virus (HBV) and its satellite hepatitis D virus (HDV). Species barriers to HBV/HDV infection are mainly determined at entry level by variations in the sequences of particular NTCP orthologs. In this study, we sought to determine whether the NTCP ortholog in woodchuck (Marmota monax), woodchuck NTCP (wNTCP) supports viral infection. We found that wNTCP is capable of supporting HBV/HDV infection in HepG2 cells, but to much lower extent than human NTCP (hNTCP), which is about 90% reduction of hNTCP. Comprehensive site-directed mutagenesis mapping of hNTCP and wNTCP revealed that the residue at position 263 is a novel site crucial for viral entry. The important role of site 263 in infection is conserved among NTCP orthologs and may therefore be a potential target for blocking the viral entry.


Assuntos
Vírus da Hepatite B/metabolismo , Vírus Delta da Hepatite/metabolismo , Especificidade de Hospedeiro/genética , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Receptores Virais/genética , Simportadores/genética , Internalização do Vírus , Sequência de Aminoácidos/genética , Animais , Sítios de Ligação/genética , Linhagem Celular Tumoral , Clonagem Molecular , Células Hep G2 , Hepatócitos/virologia , Humanos , Fígado/metabolismo , Fígado/virologia , Masculino , Marmota/virologia , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Ligação Proteica/genética , Receptores Virais/metabolismo , Simportadores/metabolismo
7.
PLoS Pathog ; 11(4): e1004840, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25902143

RESUMO

Hepatitis D virus (HDV) is the smallest virus known to infect human. About 15 million people worldwide are infected by HDV among those 240 million infected by its helper hepatitis B virus (HBV). Viral hepatitis D is considered as one of the most severe forms of human viral hepatitis. No specific antivirals are currently available to treat HDV infection and antivirals against HBV do not ameliorate hepatitis D. Liver sodium taurocholate co-transporting polypeptide (NTCP) was recently identified as a common entry receptor for HDV and HBV in cell cultures. Here we show HDV can infect mice expressing human NTCP (hNTCP-Tg). Antibodies against critical regions of HBV envelope proteins blocked HDV infection in the hNTCP-Tg mice. The infection was acute yet HDV genome replication occurred efficiently, evident by the presence of antigenome RNA and edited RNA species specifying large delta antigen in the livers of infected mice. The resolution of HDV infection appears not dependent on adaptive immune response, but might be facilitated by innate immunity. Liver RNA-seq analyses of HDV infected hNTCP-Tg and type I interferon receptor 1 (IFNα/ßR1) null hNTCP-Tg mice indicated that in addition to induction of type I IFN response, HDV infection was also associated with up-regulation of novel cellular genes that may modulate HDV infection. Our work has thus proved the concept that NTCP is a functional receptor for HDV infection in vivo and established a convenient small animal model for investigation of HDV pathogenesis and evaluation of antiviral therapeutics against the early steps of infection for this important human pathogen.


Assuntos
Hepatite D/metabolismo , Vírus Delta da Hepatite/fisiologia , Hepatócitos/metabolismo , Interações Hospedeiro-Patógeno , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Simportadores/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Antivirais/farmacologia , Células Cultivadas , Cruzamentos Genéticos , Feminino , Hepatite D/tratamento farmacológico , Hepatite D/patologia , Hepatite D/virologia , Vírus Delta da Hepatite/efeitos dos fármacos , Vírus Delta da Hepatite/imunologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Hepatócitos/virologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Imunidade Inata/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Organismos Livres de Patógenos Específicos , Simportadores/genética , Proteínas do Envelope Viral/antagonistas & inibidores , Proteínas do Envelope Viral/metabolismo
8.
J Virol ; 88(6): 3273-84, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24390325

RESUMO

UNLABELLED: The liver bile acids transporter sodium taurocholate cotransporting polypeptide (NTCP) is responsible for the majority of sodium-dependent bile salts uptake by hepatocytes. NTCP also functions as a cellular receptor for viral entry of hepatitis B virus (HBV) and hepatitis D virus (HDV) through a specific interaction between NTCP and the pre-S1 domain of HBV large envelope protein. However, it remains unknown if these two functions of NTCP are independent or if they interfere with each other. Here we show that binding of the pre-S1 domain to human NTCP blocks taurocholate uptake by the receptor; conversely, some bile acid substrates of NTCP inhibit HBV and HDV entry. Mutations of NTCP residues critical for bile salts binding severely impair viral infection by HDV and HBV; to a lesser extent, the residues important for sodium binding also inhibit viral infection. The mutation S267F, corresponding to a single nucleotide polymorphism (SNP) found in about 9% of the East Asian population, renders NTCP without either taurocholate transporting activity or the ability to support HBV or HDV infection in cell culture. These results demonstrate that molecular determinants critical for HBV and HDV entry overlap with that for bile salts uptake by NTCP, indicating that viral infection may interfere with the normal function of NTCP, and bile acids and their derivatives hold the potential for further development into antiviral drugs. IMPORTANCE: Human hepatitis B virus (HBV) and its satellite virus, hepatitis D virus (HDV), are important human pathogens. Available therapeutics against HBV are limited, and there is no drug that is clinically available for HDV infection. A liver bile acids transporter (sodium taurocholate cotransporting polypeptide [NTCP]) critical for maintaining homeostasis of bile acids serves as a functional receptor for HBV and HDV. We report here that the NTCP-binding lipopeptide that originates from the first 47 amino acids of the pre-S1 domain of the HBV L protein blocks taurocholate transport. Some bile salts dose dependently inhibit HBV and HDV infection mediated by NTCP; molecular determinants of NTCP critical for HBV and HDV entry overlap with that for bile acids transport. This work advances our understanding of NTCP-mediated HBV and HDV infection in relation to NTCP's physiological function. Our results also suggest that bile acids or their derivatives hold potential for development into novel drugs against HBV and HDV infection.


Assuntos
Vírus da Hepatite B/fisiologia , Hepatite B/metabolismo , Hepatite D/metabolismo , Vírus Delta da Hepatite/fisiologia , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Simportadores/metabolismo , Internalização do Vírus , Motivos de Aminoácidos , Transporte Biológico , Hepatite B/genética , Hepatite B/virologia , Vírus da Hepatite B/genética , Hepatite D/genética , Hepatite D/virologia , Vírus Delta da Hepatite/genética , Humanos , Transportadores de Ânions Orgânicos Dependentes de Sódio/química , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Ligação Proteica , Receptores Virais/química , Receptores Virais/genética , Receptores Virais/metabolismo , Simportadores/química , Simportadores/genética , Ácido Taurocólico/metabolismo , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
9.
J Virol ; 88(1): 237-48, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24155382

RESUMO

Severe fever with thrombocytopenia syndrome virus (SFTSV) is a novel phlebovirus in the Bunyaviridae family. Most patients infected by SFTSV present with fever and thrombocytopenia, and up to 30% die due to multiple-organ dysfunction. The mechanisms by which SFTSV enters multiple cell types are unknown. SFTSV contains two species of envelope glycoproteins, Gn (44.2 kDa) and Gc (56 kDa), both of which are encoded by the M segment and are cleaved from a precursor polypeptide (about 116 kDa) in the endoplasmic reticulum (ER). Gn fused with an immunoglobulin Fc tag at its C terminus (Gn-Fc) bound to multiple cells susceptible to the infection of SFTSV and blocked viral infection of human umbilical vein endothelial cells (HUVECs). Immunoprecipitation assays following mass spectrometry analysis showed that Gn binds to nonmuscle myosin heavy chain IIA (NMMHC-IIA), a cellular protein with surface expression in multiple cell types. Small interfering RNA (siRNA) knockdown of NMMHC-IIA, but not the closely related NMMHC-IIB or NMMHC-IIC, reduced SFTSV infection, and NMMHC-IIA specific antibody blocked infection by SFTSV but not other control viruses. Overexpression of NMMHC-IIA in HeLa cells, which show limited susceptivity to SFTSV, markedly enhanced SFTSV infection of the cells. These results show that NMMHC-IIA is critical for the cellular entry of SFTSV. As NMMHC-IIA is essential for the normal functions of platelets and human vascular endothelial cells, it is conceivable that NMMHC-IIA directly contributes to the pathogenesis of SFTSV and may be a useful target for antiviral interventions against the viral infection.


Assuntos
Febre/virologia , Cadeias Pesadas de Miosina/fisiologia , Phlebovirus/patogenicidade , Trombocitopenia/virologia , Animais , Sequência de Bases , Western Blotting , Linhagem Celular , Primers do DNA , Humanos , Microscopia Confocal , Cadeias Pesadas de Miosina/genética , Reação em Cadeia da Polimerase , RNA Interferente Pequeno/genética
10.
J Virol ; 87(14): 7977-91, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23678176

RESUMO

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.


Assuntos
Vírus da Hepatite B/metabolismo , Vírus Delta da Hepatite/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Simportadores/metabolismo , Internalização do Vírus , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Ensaio de Imunoadsorção Enzimática , Fluoresceína-5-Isotiocianato , Antígenos de Superfície da Hepatite B/metabolismo , Humanos , Camundongos , Microscopia de Fluorescência , Mutação/genética , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Precursores de Proteínas/metabolismo , Simportadores/genética , Ácido Taurocólico/metabolismo , Trítio , Células Vero
11.
J Virol ; 87(12): 7176-84, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23596296

RESUMO

Primary Tupaia hepatocytes (PTHs) are susceptible to woolly monkey hepatitis B virus (WMHBV) infection, but the identity of the cellular receptor(s) mediating WMHBV infection of PTHs remains unclear. Recently, sodium taurocholate cotransporting polypeptide (NTCP) was identified as a functional receptor for human hepatitis B virus (HBV) infection of primary human and Tupaia hepatocytes. In this study, a synthetic pre-S1 peptide from WMHBV was found to bind specifically to cells expressing Tupaia NTCP (tsNTCP) and it efficiently blocked WMHBV entry into PTHs; silencing of tsNTCP in PTHs significantly inhibited WMHBV infection. Ectopic expression of tsNTCP rendered HepG2 cells susceptible to WMHBV infection. These data demonstrate that tsNTCP is a functional receptor for WMHBV infection of PTHs. The result also indicates that NTCP's orthologs likely act as a common cellular receptor for all known primate hepadnaviruses.


Assuntos
Atelinae/virologia , Hepadnaviridae/patogenicidade , Hepatócitos/virologia , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Receptores Virais/metabolismo , Simportadores/metabolismo , Tupaia/virologia , Sequência de Aminoácidos , Animais , Células Cultivadas , Hepadnaviridae/genética , Hepadnaviridae/metabolismo , Infecções por Hepadnaviridae/virologia , Antígenos de Superfície da Hepatite B/química , Antígenos de Superfície da Hepatite B/genética , Antígenos de Superfície da Hepatite B/metabolismo , Humanos , Dados de Sequência Molecular , Precursores de Proteínas/química , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo
12.
Elife ; 1: e00049, 2012 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-23150796

RESUMO

Human hepatitis B virus (HBV) infection and HBV-related diseases remain a major public health problem. Individuals coinfected with its satellite hepatitis D virus (HDV) have more severe disease. Cellular entry of both viruses is mediated by HBV envelope proteins. The pre-S1 domain of the large envelope protein is a key determinant for receptor(s) binding. However, the identity of the receptor(s) is unknown. Here, by using near zero distance photo-cross-linking and tandem affinity purification, we revealed that the receptor-binding region of pre-S1 specifically interacts with sodium taurocholate cotransporting polypeptide (NTCP), a multiple transmembrane transporter predominantly expressed in the liver. Silencing NTCP inhibited HBV and HDV infection, while exogenous NTCP expression rendered nonsusceptible hepatocarcinoma cells susceptible to these viral infections. Moreover, replacing amino acids 157-165 of nonfunctional monkey NTCP with the human counterpart conferred its ability in supporting both viral infections. Our results demonstrate that NTCP is a functional receptor for HBV and HDV.DOI:http://dx.doi.org/10.7554/eLife.00049.001.


Assuntos
Vírus da Hepatite B/metabolismo , Vírus Delta da Hepatite/metabolismo , Hepatócitos/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Receptores Virais/metabolismo , Simportadores/metabolismo , Proteínas do Envelope Viral/metabolismo , Sequência de Aminoácidos , Animais , Transporte Biológico , Linhagem Celular , Expressão Gênica , Vírus da Hepatite B/química , Vírus da Hepatite B/genética , Vírus Delta da Hepatite/química , Vírus Delta da Hepatite/genética , Hepatócitos/patologia , Hepatócitos/virologia , Humanos , Fígado/metabolismo , Fígado/patologia , Fígado/virologia , Dados de Sequência Molecular , Transportadores de Ânions Orgânicos Dependentes de Sódio/química , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Peptídeos/química , Processos Fotoquímicos , Cultura Primária de Células , Ligação Proteica , Estrutura Terciária de Proteína , Receptores Virais/química , Receptores Virais/genética , Simportadores/química , Simportadores/genética , Ácido Taurocólico/metabolismo , Tupaia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Internalização do Vírus
13.
Elife ; 32012 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-25409679

RESUMO

Human hepatitis B virus (HBV) infection and HBV-related diseases remain a major public health problem. Individuals coinfected with its satellite hepatitis D virus (HDV) have more severe disease. Cellular entry of both viruses is mediated by HBV envelope proteins. The pre-S1 domain of the large envelope protein is a key determinant for receptor(s) binding. However, the identity of the receptor(s) is unknown. Here, by using near zero distance photo-cross-linking and tandem affinity purification, we revealed that the receptor-binding region of pre-S1 specifically interacts with sodium taurocholate cotransporting polypeptide (NTCP), a multiple transmembrane transporter predominantly expressed in the liver. Silencing NTCP inhibited HBV and HDV infection, while exogenous NTCP expression rendered nonsusceptible hepatocarcinoma cells susceptible to these viral infections. Moreover, replacing amino acids 157-165 of nonfunctional monkey NTCP with the human counterpart conferred its ability in supporting both viral infections. Our results demonstrate that NTCP is a functional receptor for HBV and HDV.


Assuntos
Vírus da Hepatite B/metabolismo , Vírus Delta da Hepatite/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Receptores Virais/metabolismo , Simportadores/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Células Cultivadas , Suscetibilidade a Doenças , Hepatite B/patologia , Hepatite B/virologia , Hepatite D/patologia , Hepatite D/virologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Hepatócitos/virologia , Humanos , Ligantes , Dados de Sequência Molecular , Transportadores de Ânions Orgânicos Dependentes de Sódio/química , Ligação Proteica , Estrutura Terciária de Proteína , Receptores Virais/química , Reprodutibilidade dos Testes , Simportadores/química , Tupaiidae , Vírion/metabolismo
14.
Cell Cycle ; 10(17): 2946-58, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21857149

RESUMO

Spinocerebellar ataxia type 10 (SCA10) is an autosomal dominant neurologic disorder, whose symptoms include cerebellar ataxia and epilepsy. The disease is caused by ATTCT expansion in the ATXN10 gene, which encodes the Ataxin-10 protein. Here we identified polo-like kinase 1 (Plk1) as one of Ataxin-10's binding partners. We show that epitope-tagged Ataxin-10 and Plk1 coimmunoprecipitate, and Plk1 phosphorylates Ataxin-10 at S77 and T82 in vitro. Knockdown of ATXN10 with siRNA in HeLa cells results in cytokinesis defects-multinucleation, which are rescued by wild-type Ataxin-10, but not the phosphor-deficient 2A mutant. Phosphorylation-specific antibodies towards pS77 detect specific signals at the midbody. Like the knockdown, overexpression of the 2A mutant generates multinucleated cells and the 2A mutant shows decreased interaction with the Plk1 polo-box domain. In addition, we found that Ataxin-10 is ubiquitinated, and is subject to proteasome-dependent degradation, which is delayed in the 2A mutant. We propose a model in which Plk1 phosphorylation of Ataxin-10 influences its degradation and cytokinesis, which may provide mechanistic insight to SCA10's pathogenesis.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Citocinese , Proteínas do Tecido Nervoso/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Ataxina-10 , Sítios de Ligação , Proteínas de Ciclo Celular/antagonistas & inibidores , Técnicas de Silenciamento de Genes , Células Gigantes/citologia , Células Gigantes/metabolismo , Células HeLa , Humanos , Fosforilação , Domínios e Motivos de Interação entre Proteínas , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Estabilidade Proteica , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Pteridinas/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Serina/metabolismo , Transdução de Sinais , Treonina/metabolismo , Transfecção , Ubiquitinação , Quinase 1 Polo-Like
15.
J Proteome Res ; 9(12): 6354-67, 2010 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-20883037

RESUMO

In recent years, electron transfer dissociation (ETD) has enjoyed widespread applications from sequencing of peptides with or without post-translational modifications to top-down analysis of intact proteins. However, peptide identification rates from ETD spectra compare poorly with those from collision induced dissociation (CID) spectra, especially for doubly charged precursors. This is in part due to an insufficient understanding of the characteristics of ETD and consequently a failure of database search engines to make use of the rich information contained in the ETD spectra. In this study, we statistically characterized ETD fragmentation patterns from a collection of 461 440 spectra and subsequently implemented our findings into pFind, a database search engine developed earlier for CID data. From ETD spectra of doubly charged precursors, pFind 2.1 identified 63-122% more unique peptides than Mascot 2.2 under the same 1% false discovery rate. For higher charged peptides as well as phosphopeptides, pFind 2.1 also consistently obtained more identifications. Of the features built into pFind 2.1, the following two greatly enhanced its performance: (1) refined automatic detection and removal of high-intensity peaks belonging to the precursor, charge-reduced precursor, or related neutral loss species, whose presence often set spectral matching askew; (2) a thorough consideration of hydrogen-rearranged fragment ions such as z + H and c - H for peptide precursors of different charge states. Our study has revealed that different charge states of precursors result in different hydrogen rearrangement patterns. For a fragment ion, its propensity of gaining or losing a hydrogen depends on (1) the ion type (c or z) and (2) the size of the fragment relative to the precursor, and both dependencies are affected by (3) the charge state of the precursor. In addition, we discovered ETD characteristics that are unique for certain types of amino acids (AAs), such as a prominent neutral loss of SCH(2)CONH(2) (90.0014 Da) from z ions with a carbamidomethylated cysteine at the N-terminus and a neutral loss of histidine side chain C(4)N(2)H(5) (81.0453 Da) from precursor ions containing histidine. The comprehensive list of ETD characteristics summarized in this paper should be valuable for automated database search, de novo peptide sequencing, and manual spectral validation.


Assuntos
Espectrometria de Massas/métodos , Peptídeos/análise , Proteômica/métodos , Sequência de Aminoácidos , Transporte de Elétrons , Dados de Sequência Molecular , Peptídeos/química , Fosfopeptídeos/análise , Fosfopeptídeos/química , Reprodutibilidade dos Testes
16.
Proc Natl Acad Sci U S A ; 107(41): 17639-44, 2010 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-20876089

RESUMO

The mammalian oocyte possesses powerful reprogramming factors, which can reprogram terminally differentiated germ cells (sperm) or somatic cells within a few cell cycles. Although it has been suggested that use of oocyte-derived transcripts may enhance the generation of induced pluripotent stem cells, the reprogramming factors in oocytes are undetermined, and even the identified proteins composition of oocytes is very limited. In the present study, 7,000 mouse oocytes at different developmental stages, including the germinal vesicle stage, the metaphase II (MII) stage, and the fertilized oocytes (zygotes), were collected. We successfully identified 2,781 proteins present in germinal vesicle oocytes, 2,973 proteins in MII oocytes, and 2,082 proteins in zygotes through semiquantitative MS analysis. Furthermore, the results of the bioinformatics analysis indicated that different protein compositions are correlated with oocyte characteristics at different developmental stages. For example, specific transcription factors and chromatin remodeling factors are more abundant in MII oocytes, which may be crucial for the epigenetic reprogramming of sperm or somatic nuclei. These results provided important knowledge to better understand the molecular mechanisms in early development and may improve the generation of induced pluripotent stem cells.


Assuntos
Diferenciação Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Oócitos/crescimento & desenvolvimento , Proteoma/genética , Zigoto/metabolismo , Animais , Biologia Computacional , DNA/metabolismo , Epigênese Genética/genética , Espectrometria de Massas , Camundongos , Oócitos/metabolismo , Proteômica/métodos , Transdução de Sinais/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...