Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
J Mol Graph Model ; 102: 107763, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33069124

RESUMO

The molecular electronic density theory (MEDT) was invested to elucidate the chemo-, regio- and stereo-selectivity of the 1,3-dipolar cycloaddition between Diazomethane (DZM) and Psilostachyin (PSH). The DFT method at B3LYP/6-31 + G (d,p) level of theory was used. Reactivity indices, transition structures theory, IGM and ELF analysis were employed to reveal the mechanism of the reaction. The addition of DZM to PSH takes place through a one-step mechanism and an asynchronous transition states. Eight possible addition channels of reaction were investigated (addition of C (sp2) to Diazomethane at C4, C5, C6 or C7). The addition of C (sp2) at C5 leading to P1 product is the preferred channel. The addition of ether does not affect the chemo-, regio- and stereo-selectivity of the reaction. Analysis of transfer of charges along the IRC path associated with the P1 product shows a polar character for the studied reaction. We have also used the noncovalent interaction (NCI) which is very helpful to reveal the most favored addition channel of the reaction, by analyzing the weak interactions in different TSs. Finally, we investigate about the potential of inhibition of some pyrazoline compounds against COVID-19-Mpro by performing a molecular docking calculations.


Assuntos
Antivirais/química , Antivirais/farmacologia , Lactonas/química , Lactonas/farmacologia , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/enzimologia , Sesquiterpenos/química , Sesquiterpenos/farmacologia , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/efeitos dos fármacos , COVID-19/virologia , Reação de Cicloadição , Diazometano/química , Humanos , Simulação de Acoplamento Molecular , Pandemias , Inibidores de Proteases/química , Inibidores de Proteases/farmacologia , Conformação Proteica , Pirazóis/química , Pirazóis/farmacologia , Eletricidade Estática , Tratamento Farmacológico da COVID-19
2.
Photodiagnosis Photodyn Ther ; 30: 101801, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32360854

RESUMO

Nasopharyngeal carcinoma (NPC) is of high prevalence in Hong Kong and southern China. The pathogenesis of NPC is closely associated with Epstein-Barr virus (EBV) infection via regulation of viral oncoprotein latent membrane protein 1 (LMP1). The conventional treatment for NPC is chemo-radiotherapy, but the prognosis remains poor for advanced stage, recurrent and metastatic NPC. Photodynamic therapy (PDT) is a therapeutic approach to combat tumors. PDT effectiveness depends on the interaction of photosensitizers, light and molecular oxygen. 5- aminolevulinic acid hexyl derivative (H-ALA) is one of the photosensitizers derived from 5-ALA. H-ALA with improved lipophilic properties by adding a long lipophilic chain (hexyl group) to 5-ALA, resulted in better penetration into cell cytoplasm. In this study, the effect of H-ALA-PDT on NPC cells (EBV positive C666-1 and EBV negative CNE2) was investigated. The H-ALA mediated cellular uptake and cytotoxicity was revealed via flow cytometry analysis and MTT assay respectively. H-ALA PDT mediated protein modulation was analysed by western blot analysis. Our finding reported that the cellular uptake of H-ALA in C666-1 and CNE2 cells was in a time dependent manner. H-ALA PDT was effective to C666-1 and CNE2 cells. EBV LMP1 proteins was expressed in C666-1 cells only and its expression was responsive to H-ALA PDT in a dose dependent manner. This work revealed the potential of H-ALA PDT as a treatment regiment for EBV positive NPC cells. Understanding the mechanism of H-ALA mediated PDT could develop improved strategies for the treatment of NPC.


Assuntos
Ácido Aminolevulínico/análogos & derivados , Terapia com Luz de Baixa Intensidade/métodos , Neoplasias Nasofaríngeas/tratamento farmacológico , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia , Proteínas da Matriz Viral/efeitos dos fármacos , Ácido Aminolevulínico/farmacocinética , Ácido Aminolevulínico/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular , Herpesvirus Humano 4/efeitos dos fármacos , Humanos , Lasers Semicondutores/uso terapêutico , Fármacos Fotossensibilizantes/farmacocinética , Protoporfirinas/farmacocinética
3.
Microbiol Immunol ; 62(1): 34-43, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29210473

RESUMO

M2 protein of influenza A virus is a proton channel spanning the viral envelope. Activity of this proton channel is required for uncoating of viral particles and equilibrating the pH across the trans Golgi apparatus, which prevents conformational change in hemagglutinin. Amantadine, an anti-influenza A virus drug, inhibits M2 proton channel activity by binding to the channel pore; however, most currently circulating influenza A viruses are amantadine-resistant. The most prevalent resistant mutation is a substitution from Ser31 to Asn31 in M2. Further atomistic analysis of ligand-M2 complexes is needed to provide new approaches for the design of novel M2 channel blockers. Here, the free energy profiles of the binding kinetics of M2 channel blockers were examined by well-tempered metadynamics simulations and it was found that amantadine first binds to Asp24 of S31 M2 and forms a metastable conformation. In contrast, the free energy profiles of adamantyl bromothiophene dual inhibitor with either S31 M2 or N31 M2 are broad funnel-shaped curves, suggesting that adamantyl bromothiophene does not form metastable complexes with M2. The trajectory of well-tempered metadynamics simulations revealed that steric hindrance between adamantyl bromothiophene and S31 M2 interrupts formation of a metastable conformation at Asp24 and that a halogen bond between the bromine atom and N31 is responsible for pulling down the ligand to the channel pore of N31 M2 in the absence of a metastable state. Binding pathways of M2 channel blockers to M2 are here proposed on the basis of these findings; they may provide new approaches to designing further M2 channel blockers.


Assuntos
Vírus da Influenza A/metabolismo , Simulação de Dinâmica Molecular , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/efeitos dos fármacos , Amantadina/farmacologia , Antivirais/farmacologia , Sítios de Ligação , Farmacorresistência Viral/efeitos dos fármacos , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/genética , Cinética , Ligantes , Mutação , Ressonância Magnética Nuclear Biomolecular , Conformação Proteica/efeitos dos fármacos , Proteínas da Matriz Viral/genética , Água/química
4.
Antiviral Res ; 134: 77-88, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27565992

RESUMO

The spiro compound 5,6-dimethyl-3H,3'H-spiro(benzofuran-2,1'-isobenzofuran)-3,3'-dione (KR-23502) has antiviral activity against influenza A and more potently B viruses. The aim of this study is to elucidate its mechanism of action. Subcellular localization and time-course expression of influenza B viral proteins, nucleoprotein (NP) and matrix protein 1 (M1), showed that KR-23502 reduced their amounts within 5 h post-infection. Early steps of virus life cycle, including virus entry, nuclear localization of NP and viral RNA-dependent RNA replication, were not affected by KR-23502. Instead it interrupted a later event corresponding to nuclear export of NP and M1 proteins. Delivery of viral ribonucleoprotein (vRNP)-M1 complex has been known to be mediated by the viral nuclear export protein (NEP) through interaction with cellular chromosomal maintenance 1 (CRM1) protein. In this study, we experimentally demonstrated that the compound targets the nuclear export of vRNP. Moreover, a single mutation (aspartate to glycine) at amino acid position 54 in M1 [M1(D54G)] was detected after 18 passages in the presence of KR-23502 with a 2-fold increase in 50% effective concentration indicating that this compound has a relatively high genetic barrier to resistance. Interestingly, it was observed that proteasome-mediated degradation of M1(D54G) was attenuated by KR-23502. In conclusion, we suggest that KR-23502 shows its anti-influenza activity by downregulating NEP/CRM1-mediated nuclear export of influenza vRNP and M1. KR-23502 provides a core chemical skeleton for further structure-based design of novel antivirals against influenza viruses.


Assuntos
Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Antivirais/farmacologia , Benzofuranos/farmacologia , Vírus da Influenza B/efeitos dos fármacos , Ribonucleoproteínas/efeitos dos fármacos , Proteínas da Matriz Viral , Núcleo Celular/metabolismo , Humanos , Vírus da Influenza B/química , Vírus da Influenza B/genética , Influenza Humana/tratamento farmacológico , Mutação , RNA Viral , Ribonucleoproteínas/química , Ribonucleoproteínas/genética , Proteínas da Matriz Viral/efeitos dos fármacos , Proteínas Virais/metabolismo , Replicação Viral/efeitos dos fármacos
5.
Adv Exp Med Biol ; 827: 205-26, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25387967

RESUMO

The influenza A virus matrix protein 2 (M2 protein) is a pH-regulated proton channel embedded in the viral membrane. Inhibition of the M2 proton channel has been used to treat influenza infections for decades due to the crucial role of this protein in viral infection and replication. However, the widely-used M2 inhibitors, amantadine and rimantadine, have gradually lost their efficiencies because of naturally-occurring drug resistant mutations. Therefore, investigation of the structure and function of the M2 proton channel will not only increase our understanding of this important biological system, but also lead to the design of novel and effective anti-influenza drugs. Despite the simplicity of the M2 molecular structure, the M2 channel is highly flexible and there have been controversies and arguments regarding the channel inhibition mechanism and the proton conduction mechanism. In this book chapter, we will first carefully review the experimental and computational studies of the two possible drug binding sites on the M2 protein and explain the mechanisms regarding how inhibitors prevent proton conduction. Then, we will summarize our recent molecular dynamics simulations of the drug-resistant mutant channels and propose mechanisms for drug resistance. Finally, we will discuss two existing proton conduction mechanisms and talk about the remaining questions regarding the proton-relay process through the channel. The studies reviewed here demonstrate how molecular modeling and simulations have complemented experimental work and helped us understand the M2 channel structure and function.


Assuntos
Prótons , Proteínas da Matriz Viral/efeitos dos fármacos , Conformação Proteica , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/metabolismo
6.
Curr Pharm Des ; 17(17): 1720-39, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21619529

RESUMO

While the seasonal influenza viruses spreading around the world cause the annual epidemics, the recent outbreaks of influenza A virus subtype H5N1 and pandemic H1N1 have raised a global human health concerns. In this review, the applicability of computational techniques focused on three important targets in the viral life cycle: hemagglutinin, neuraminidase and M2 proton channel are summarized. Protein mechanism of action, substrate binding specificity and drug resistance, ligand-target interactions of substrate/inhibitor binding to these three proteins either wild-type or mutant strains are discussed and compared. Advances on the novel anti-influenza agents designed specifically to combat the avian H5N1 and pandemic H1N1 viruses are introduced. A better understanding of molecular inhibition and source of drug resistance as well as a set of newly designed compounds is greatly useful as a rotational guide for synthetic and medicinal chemists to develop a new generation of anti-influenza drugs.


Assuntos
Antivirais/farmacologia , Hemaglutininas Virais/efeitos dos fármacos , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Virus da Influenza A Subtipo H5N1/efeitos dos fármacos , Neuraminidase/efeitos dos fármacos , Proteínas da Matriz Viral/efeitos dos fármacos , Biologia Computacional , Vírus da Influenza A Subtipo H1N1/enzimologia , Vírus da Influenza A Subtipo H1N1/metabolismo , Virus da Influenza A Subtipo H5N1/enzimologia , Virus da Influenza A Subtipo H5N1/metabolismo
8.
Biochim Biophys Acta ; 1808(2): 547-53, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20599692

RESUMO

The recent outbreaks of avian flu in Southeast Asia and swine flu in Mexico City painfully exemplify the ability of the influenza virus to rapidly mutate and develop resistance to modern medicines. This review seeks to detail the molecular mechanism by which the influenza virus has obtained resistance to amino-adamantyls, one of only two classes of drugs that combat the flu. Amino-adamantyls target the viral M2 H(+) channel and have become largely ineffective due to mutations in the transmembrane domain of the protein. Herein we describe these resistance rendering mutations and the compounded effects they have upon the protein's function and resulting virus viability.


Assuntos
Adamantano/farmacologia , Influenza Humana/tratamento farmacológico , Amantadina/farmacologia , Sequência de Aminoácidos , Farmacorresistência Viral/genética , Genes Virais , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Glicoproteínas de Hemaglutininação de Vírus da Influenza/efeitos dos fármacos , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Humanos , Vírus da Influenza A/química , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/genética , Vírus da Influenza A/patogenicidade , Influenza Humana/virologia , Canais Iônicos/química , Canais Iônicos/efeitos dos fármacos , Canais Iônicos/genética , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Estabilidade Proteica , Estrutura Terciária de Proteína , Rimantadina/farmacologia , Homologia de Sequência de Aminoácidos , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/efeitos dos fármacos , Proteínas da Matriz Viral/genética
9.
Molecules ; 15(9): 6127-39, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20877211

RESUMO

Epstein-Barr virus (EBV)-encoded latent membrane protein 1 (LMP1) has been known to have oncogenic properties during latent infection in nasopharyngeal carcinoma (NPC). Genetic manipulation of LMP1 expression may provide a novel strategy for the treatment of NPC. DNAzymes are synthetic, single-stranded DNA catalysts that can be engineered to bind and cleave the target mRNA of a disease-causing gene. By targeting the LMP1 mRNA, we successfully obtained a phosphorothioate-modified ''10-23'' DNAzyme namely DZ1, through screening a series of DNAzymes. DZ1 could significantly down-regulate the expression of LMP1 in NPC cells, inhibit cell proliferation, metastasis, promote apoptosis and enhance radiosensitivity of NPC through interfering signal pathways which are abnormally activated by LMP1, including NF-κB, AP-1 and STAT3 signal pathways. Together, interfering LMP1 signaling pathway could be a promising strategy to target the malignant phenotypes of NPC.


Assuntos
DNA Catalítico/farmacologia , Genes Virais/efeitos dos fármacos , Herpesvirus Humano 4/efeitos dos fármacos , Neoplasias Nasofaríngeas/tratamento farmacológico , Proteínas da Matriz Viral/antagonistas & inibidores , DNA Catalítico/química , DNA Catalítico/uso terapêutico , Neoplasias Nasofaríngeas/virologia , Transdução de Sinais/efeitos dos fármacos , Proteínas da Matriz Viral/efeitos dos fármacos , Proteínas da Matriz Viral/genética
10.
Proc Natl Acad Sci U S A ; 107(2): 872-7, 2010 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-20080768

RESUMO

Epstein-Barr virus (EBV) is associated with a variety of human tumors. Although the EBV-infected normal B cells in vitro and the EBV-carrying B cell lymphomas in immunodeficient patients express the full set of latent proteins (type III latency), the majority of EBV-associated malignancies express the restricted type I (EBNA-1 only) or type II (EBNA-1 and LMPs) viral program. The mechanisms responsible for these different latent viral gene expression patterns are only partially known. IL-21 is a potent B cell activator and plasma cell differentiation-inducer cytokine produced by CD4(+) T cells. We studied its effect on EBV-carrying B cells. In type I Burkitt lymphoma (BL) cell lines and in the conditional lymphoblastoid cell line (LCL) ER/EB2-5, IL-21 potently activated STAT3 and induced the expression of LMP-1, but not EBNA-2. The IL-21-treated type I Jijoye M13 BL line ceased to proliferate, and this was paralleled by the induction of IRF4 and the down-regulation of BCL6 expression. In the type III LCLs and BL lines, IL-21 repressed the C-promoter-derived and LMP-2A mRNAs, whereas it up-regulated the expression of LMP-1 mRNAs. The IL-21-treated type III cells underwent plasma cell differentiation with the induction of Blimp-1, and high levels of Ig and Oct-2. IL-21 might be involved in the EBNA-2-independent expression of LMP-1 in EBV-carrying type II cells. In light of the fact that IL-21 is already in clinical trials for the treatment of multiple malignancies, the in vivo modulation of EBV gene expression by IL-21 might have therapeutic benefits for the EBV-carrying malignancies.


Assuntos
Linfócitos B/imunologia , Linfócitos B/virologia , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Herpesvirus Humano 4/genética , Interleucinas/farmacologia , Proteínas da Matriz Viral/genética , Linfócitos B/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Antígenos Nucleares do Vírus Epstein-Barr/farmacologia , Genoma Viral , Humanos , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/efeitos dos fármacos , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Linfoma de Células B/imunologia , Linfoma de Células B/patologia , Linfoma de Células B/virologia , Neoplasias/genética , Neoplasias/virologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas da Matriz Viral/efeitos dos fármacos , Proteínas da Matriz Viral/farmacologia , Proteínas Virais/farmacologia , Latência Viral/genética
11.
Protein Cell ; 1(3): 246-58, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21203971

RESUMO

The M2 proteins of influenza A and B virus, AM2 and BM2, respectively, are transmembrane proteins that oligomerize in the viral membrane to form proton-selective channels. Proton conductance of the M2 proteins is required for viral replication; it is believed to equilibrate pH across the viral membrane during cell entry and across the trans-Golgi membrane of infected cells during viral maturation. In addition to the role of M2 in proton conductance, recent mutagenesis and structural studies suggest that the cytoplasmic domains of the M2 proteins also play a role in recruiting the matrix proteins to the cell surface during virus budding. As viral ion channels of minimalist architecture, the membrane-embedded channel domain of M2 has been a model system for investigating the mechanism of proton conduction. Moreover, as a proven drug target for the treatment of influenza A infection, M2 has been the subject of intense research for developing new anti-flu therapeutics. AM2 is the target of two anti-influenza A drugs, amantadine and rimantadine, both belonging to the adamantane class of compounds. However, resistance of influenza A to adamantane is now widespread due to mutations in the channel domain of AM2. This review summarizes the structure and function of both AM2 and BM2 channels, the mechanism of drug inhibition and drug resistance of AM2, as well as the development of new M2 inhibitors as potential anti-flu drugs.


Assuntos
Vírus da Influenza A/química , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza B/química , Vírus da Influenza B/efeitos dos fármacos , Proteínas da Matriz Viral/química , Proteínas Virais/química , Amantadina/farmacologia , Antivirais/farmacologia , Farmacorresistência Viral/genética , Genes Virais , Humanos , Vírus da Influenza A/genética , Vírus da Influenza B/genética , Canais Iônicos/química , Canais Iônicos/efeitos dos fármacos , Canais Iônicos/genética , Modelos Moleculares , Mutação , Estrutura Terciária de Proteína , Rimantadina/farmacologia , Proteínas da Matriz Viral/efeitos dos fármacos , Proteínas da Matriz Viral/genética , Proteínas Virais/efeitos dos fármacos , Proteínas Virais/genética
12.
J Leukoc Biol ; 87(1): 165-72, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19801502

RESUMO

APCs operate frequently under oxidative stress induced by aging, tissue damage, pathogens, or inflammatory responses. Phagocytic cells produce peroxides and free-radical species that facilitate pathogen clearance and can in the case of APCs, also lead to oxidative modifications of antigenic proteins and peptides. Little information is available presently about the consequences of such modifications on the immune response. To model oxidative modification of an immunodominant antigenic peptide, we oxidized the methionine residue of the human CMV pp65(495-503) (NLVPMVATV) peptide. Such modifications of an antigenic peptide can affect MHC binding or TCR recognition. Using binding and dissociation assays, we demonstrate that oxidative modification of the CMVpp65(495-503) peptide leads to a decreased binding of the pMHC complex to the TCR, whereas binding of the peptide to the MHC class I molecule is not impaired. Additionally, we show that CD8(+) T cells have a decreased proliferation and IFN-gamma production when stimulated with oxidized CMVpp65(495-503) peptide. Spectratyping the antigen-binding site of the TCR of responding T cells demonstrates that the CMVpp65(495-503) and the CMVoxpp65(495-503) peptides preferentially stimulate BV8 T cells. Sequencing of this dominant BV family reveals a highly conserved CDR3 amino acid motif, independent of the mode of stimulation, demonstrating the recruitment of the same T cell clonotypes. Our results suggest that oxidative modification of antigenic peptides may affect T cell responses severely by binding T cell clones with different affinity. This may lead to an altered immune response against infectious agents as well as against tumor or autoantigens under oxidative stress conditions.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Epitopos Imunodominantes/imunologia , Estresse Oxidativo/imunologia , Fosfoproteínas/imunologia , Proteínas da Matriz Viral/imunologia , Adulto , Motivos de Aminoácidos , Antígenos Virais/química , Antígenos Virais/efeitos dos fármacos , Células Cultivadas/imunologia , Infecções por Citomegalovirus/imunologia , Feminino , Antígenos HLA-A/imunologia , Antígeno HLA-A2 , Humanos , Peróxido de Hidrogênio/farmacologia , Epitopos Imunodominantes/efeitos dos fármacos , Interferon gama/biossíntese , Ativação Linfocitária , Masculino , Metionina/química , Oxirredução , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/efeitos dos fármacos , Fragmentos de Peptídeos/imunologia , Fosfoproteínas/química , Fosfoproteínas/efeitos dos fármacos , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/efeitos dos fármacos
13.
Biochem Biophys Res Commun ; 388(2): 413-7, 2009 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-19665993

RESUMO

The M2 proton channel is one of indispensable components for the influenza A virus that plays a vital role in its life cycle and hence is an important target for drug design against the virus. In view of this, the three-dimensional structure of the H1N1-M2 channel was developed based on the primary sequence taken from a patient recently infected by the H1N1 (swine flu) virus. With an explicit water-membrane environment, molecular docking studies were performed for amantadine and rimantadine, the two commercial drugs generally used to treat influenza A infection. It was found that their binding affinity to the H1N1-M2 channel is significantly lower than that to the H5N1-M2 channel, fully consistent with the recent report that the H1N1 swine virus was resistant to the two drugs. The findings and the relevant analysis reported here might provide useful structural insights for developing effective drugs against the new swine flu virus.


Assuntos
Adamantano/química , Antivirais/química , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Rimantadina/química , Proteínas da Matriz Viral/química , Adamantano/farmacologia , Sequência de Aminoácidos , Animais , Antivirais/farmacologia , Desenho de Fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica , Rimantadina/farmacologia , Proteínas da Matriz Viral/efeitos dos fármacos
14.
Tidsskr Nor Laegeforen ; 128(22): 2601-6, 2008 Nov 20.
Artigo em Norueguês | MEDLINE | ID: mdl-19023375

RESUMO

BACKGROUND: Influenza virus infection can be prevented and treated with antiviral drugs. The usage of such drugs in Norway has been infrequent, however, they are an important component in our pandemic preparedness planning, as it will probably be difficult to get access to the appropriate vaccine in time before the pandemic reaches the country. The first generation of influenza drugs acquired resistance to a large degree, in contrast to the modern neuraminidase inhibitors that until recently have had minor problems with resistance. MATERIAL AND METHODS: This review is based on research found in relevant published literature, together with experience from a virology reference laboratory and participation in a national and international surveillance including susceptibility testing. RESULTS AND INTERPRETATION: While resistance has been a longstanding problem with the use of the "old" influenza drugs amantadine and rimantadine, only during the winter 2007/2008 did it become clear, that a certain type of virus acquired widespread resistance against the neuraminidase inhibitor oseltamivir. Resistance surveillance is crucial for the correct choice of empiric treatment for influenza infection, and will be one of the most important tasks at the National Influenza Centre in certain phases of a pandemic. The current situation with an increasing resistance problem strengthens the need to conduct continuous monitoring of antiviral susceptibility, as well as development of new antiviral drugs and treatment regimes.


Assuntos
Farmacorresistência Viral , Influenza Humana/tratamento farmacológico , Animais , Antivirais/efeitos adversos , Antivirais/uso terapêutico , Aves , Controle de Doenças Transmissíveis , Surtos de Doenças/prevenção & controle , Farmacorresistência Viral/genética , Inibidores Enzimáticos/uso terapêutico , Humanos , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/genética , Vírus da Influenza B/efeitos dos fármacos , Vírus da Influenza B/genética , Influenza Aviária/transmissão , Influenza Aviária/virologia , Gammainfluenzavirus/efeitos dos fármacos , Gammainfluenzavirus/genética , Cooperação Internacional , Neuraminidase/antagonistas & inibidores , Orthomyxoviridae/efeitos dos fármacos , Orthomyxoviridae/genética , Proteínas da Matriz Viral/efeitos dos fármacos , Zoonoses/virologia
15.
Bioorg Med Chem Lett ; 18(23): 6156-60, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18947998

RESUMO

The new 2-alkyl-2-aminoadamantanes and analogues 4-10 were designed and synthesized by simplification of the structure of the potent anti-influenza virus A spiranic aminoadamantane heterocycles 2 and 3. The aim of the present work was to examine the effects of bulky and extended lipophilic moieties attached to amantadine 1 on binding to the M2 channel and the resulting antiviral potency. The binding affinities of the compounds to the M2 protein of influenza virus A/chicken/Germany/27 (Weybridge strain; H7N7) were measured for the first time using an assay based on quenching of Trp-41 fluorescence by His-37 protonation, and their antiviral potencies were evaluated against the replication of influenza virus A H2N2 and H3N2 subtypes and influenza virus B in MDCK cells. Of the various 2-alkyl-2-aminoadamantanes, and analogues, spiro[piperidine-2,2'-adamantane] 3 had the strongest M2 binding and antiviral potency, which were similar those of amantadine 1. The relative binding affinities suggested that the rigid carbon framework provided by the pyrrolidine or piperidine rings results in a more favorable orientation inside the M2 channel pore as compared to large, freely rotating alkyl groups. The aminoadamantane derivatives exhibited similar NMDA antagonistic activity to amantadine 1. A striking finding was the antiviral activity of the adamantanols 4, and 6, which lack any NMDA antagonist activity.


Assuntos
Adamantano , Vírus da Influenza A/efeitos dos fármacos , N-Metilaspartato/antagonistas & inibidores , Proteínas da Matriz Viral/efeitos dos fármacos , Adamantano/análogos & derivados , Adamantano/síntese química , Adamantano/química , Adamantano/farmacologia , Amantadina/farmacologia , Antivirais/química , Antivirais/farmacologia , Estrutura Molecular , Relação Estrutura-Atividade
16.
Proc Natl Acad Sci U S A ; 105(5): 1483-8, 2008 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-18230730

RESUMO

The M2 protein of influenza A virus forms a transmembrane proton channel important for viral infection and replication. Amantadine blocks this channel, thus inhibiting viral replication. Elucidating the high-resolution structure of the M2 protein and its change upon amantadine binding is crucial for designing antiviral drugs to combat the growing resistance of influenza A viruses against amantadine. We used magic-angle-spinning solid-state NMR to determine the conformation and dynamics of the transmembrane domain of the protein M2TMP in the apo- and amantadine-bound states in lipid bilayers. (13)C chemical shifts and torsion angles of the protein in 1,2-dilauroyl-sn-glycero-3-phosphatidylcholine (DLPC) bilayers indicate that M2TMP is alpha-helical in both states, but the average conformation differs subtly, especially at the G34-I35 linkage and V27 side chain. In the liquid-crystalline membrane, the complexed M2TMP shows dramatically narrower lines than the apo peptide. Analysis of the homogeneous and inhomogeneous line widths indicates that the apo-M2TMP undergoes significant microsecond-time scale motion, and amantadine binding alters the motional rates, causing line-narrowing. Amantadine also reduces the conformational heterogeneity of specific residues, including the G34/I35 pair and several side chains. Finally, amantadine causes the helical segment N-terminal to G34 to increase its tilt angle by 3 degrees , and the G34-I35 torsion angles cause a kink of 5 degrees in the amantadine-bound helix. These data indicate that amantadine affects the M2 proton channel mainly by changing the distribution and exchange rates among multiple low-energy conformations and only subtly alters the average conformation and orientation. Amantadine-resistant mutations thus may arise from binding-incompetent changes in the conformational equilibrium.


Assuntos
Amantadina/farmacologia , Antivirais/farmacologia , Proteínas da Matriz Viral/efeitos dos fármacos , Humanos , Ressonância Magnética Nuclear Biomolecular/métodos , Conformação Proteica/efeitos dos fármacos , Prótons , Proteínas da Matriz Viral/química
17.
Bioorg Med Chem Lett ; 17(14): 3947-52, 2007 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-17502147

RESUMO

(19)F NMR probes were used to follow interactions between ligands in the aminoadamantane series, amantadine (Am) 1 and 3-F-Am 2, and the 5-F-Trp20 transmembrane fragment of the influenza A M2 proton channel (F-M2TM 3) in dodecylphosphocholine micelles over the pH range 5-8. Above pH 7, when the peptide adopts a tetrameric state that is able to bind channel blocking ligands, (19)F-Trp signals from both the free and bound states of the M2TM tetramer are resolved. This differentiation of bound and unbound states of the M2TM receptor by (19)F NMR may provide a system for SAR studies.


Assuntos
Amantadina/farmacologia , Antivirais/farmacologia , Espectroscopia de Ressonância Magnética/métodos , Micelas , Proteínas da Matriz Viral/efeitos dos fármacos , Flúor , Concentração de Íons de Hidrogênio , Relação Estrutura-Atividade , Proteínas da Matriz Viral/metabolismo
18.
Braz J Med Biol Res ; 39(5): 677-85, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16648906

RESUMO

Nasopharyngeal carcinoma (NPC) is notorious for the metastases, which are in close association with Epstein-Barr virus-encoded latent membrane protein 1 (LMP1). Arsenic trioxide (As2O3) has been shown to induce apoptosis and differentiation in NPC xenografts. Then, can it repress the cancer cells' metastasis potential? To elucidate this issue, the present study was performed. LMP1-negative cell line HNE1 and LMP1-positive cell line HNE1-LMP1 were used as in vitro model. Cells (1 x 10(5)/mL) were cultured with or without 3 microM As2O3 for 48 h. Then the survival cells were collected to investigate their potential of colony formation, attachment, invasion, and migration. Both confocal immunofluorescence staining and Western blot were used to detect the changes of LMP1 expression. The changes of MMP-9 were examined by RT-PCR assay and Western blot. The results were as follow: i) the colony formation inhibition rate (75.41 +/- 3.9% in HNE1-LMP1 cells vs 37.89 +/- 4.9% in HNE1 cells), the rate of attachment (HNE1-LMP1 vs HNE1: 56.40 +/- 3.5 vs 65.87 +/- 5.9%), the invasion inhibitory rate (HNE1-LMP1 vs HNE1: 56.50 +/- 3.7 and 27.91 +/- 2.1%), and the migration inhibitory rate (HNE1-LMP1 vs HNE1: 48.70 +/- 3.9 vs 29.19 +/- 6.27%) were all significantly different between the two cell lines (P < 0.01). ii) LMP1 was down-regulated in As2O3-treated HNE1-LMP1 cells. iii) The reduction of MMP-9 was found in As2O3-treated groups, more evident in HNE1-LMP1 cells. Thus, we conclude that As2O3 can reduce metastasis potential of NPC cells, involving inhibition of MMP-9 expression. LMP1 were also reduced in this process and seemed to enhance anti-metastasis activity of As2O3.


Assuntos
Antineoplásicos/farmacologia , Arsenicais/farmacologia , Metaloproteinase 9 da Matriz/efeitos dos fármacos , Neoplasias Nasofaríngeas/tratamento farmacológico , Óxidos/farmacologia , Proteínas da Matriz Viral/efeitos dos fármacos , Trióxido de Arsênio , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Imunofluorescência , Regulação Neoplásica da Expressão Gênica , Humanos , Metaloproteinase 9 da Matriz/genética , Microscopia Confocal , Neoplasias Nasofaríngeas/patologia , Invasividade Neoplásica/patologia , Metástase Neoplásica/patologia , RNA Mensageiro/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas da Matriz Viral/genética
19.
Tumori ; 92(2): 140-8, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16724694

RESUMO

AIMS AND BACKGROUND: It was documented that nasopharyngeal carcinoma (NPC) is associated with Epstein-Barr virus (EBV) and that EBV-encoded latent membrane protein-1 expression (LMP1) plays an important role in the pathogenesis of NPC. In preclinical studies, arsenic trioxide (As2O3) has been identified as a promising anticancer agent for treatment of NPC. The purpose of this study is to investigate if this agent can inhibit the expression of LMP1 and therefore lead to growth inhibition of NPC cells in vitro. METHODS: LMP1-positive NPC cells, HNE1-LMP1, were treated with 3 micromol/L of As2O3 for 96 hours. The LMP1 protein expression and mRNA level in HNE1-LMP1 cells were determined by western blot, confocal immunofluorescence staining and semiquantitative reverse transcriptase reaction (RT-PCR). Apoptosis was determined by light microscopy and the TUNEL method. Alterations in the cell cycle distribution were also investigated by flow cytometry. MTT assay and colony formation assay were used to detect the proliferation of the cells. The LMP1-negative parental cell lines HNE1 and HNE2 were used as control in an attempt to elucidate the role of LMP1 in the anticancer effect of As2O3 on NPC cells. RESULTS: The expression of LMP1 at the protein and mRNA level was reduced after exposure to 3 micromol/L As2O3. This dose of As2O3 significantly induced apoptosis and growth retardation of HNE1-LMP1 cells. In addition, more HNE1-LMP1 cells were induced to G0/G1 and G2/M arrest. The same dose of As2O3 had a moderate effect on HNE1 and HNE2 cells. CONCLUSION: Arsenic trioxide can inhibit LMP1 expression and dictate apoptosis and alterations of cell cycle distribution as well as growth retardation. LMP1-positive NPC cells are more sensitive to As2O3 treatment than LMP1-negative NPC cells.


Assuntos
Antineoplásicos/farmacologia , Arsenicais/farmacologia , Neoplasias Nasofaríngeas/tratamento farmacológico , Neoplasias Nasofaríngeas/metabolismo , Óxidos/farmacologia , Proteínas da Matriz Viral/efeitos dos fármacos , Proteínas da Matriz Viral/metabolismo , Apoptose , Trióxido de Arsênio , Western Blotting , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Citometria de Fluxo , Imunofluorescência , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inibidores do Crescimento/farmacologia , Humanos , Marcação In Situ das Extremidades Cortadas , Neoplasias Nasofaríngeas/patologia , Proteínas Oncogênicas Virais/efeitos dos fármacos , Proteínas Oncogênicas Virais/metabolismo , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ensaio Tumoral de Célula-Tronco , Proteínas da Matriz Viral/genética
20.
Braz. j. med. biol. res ; 39(5): 677-685, May 2006. ilus, graf
Artigo em Inglês | LILACS | ID: lil-425788

RESUMO

Nasopharyngeal carcinoma (NPC) is notorious for the metastases, which are in close association with Epstein-Barr virus-encoded latent membrane protein 1 (LMP1). Arsenic trioxide (As2O3) has been shown to induce apoptosis and differentiation in NPC xenografts. Then, can it repress the cancer cells' metastasis potential? To elucidate this issue, the present study was performed. LMP1-negative cell line HNE1 and LMP1-positive cell line HNE1-LMP1 were used as in vitro model. Cells (1 x 10(5)/mL) were cultured with or without 3 æM As2O3 for 48 h. Then the survival cells were collected to investigate their potential of colony formation, attachment, invasion, and migration. Both confocal immunofluorescence staining and Western blot were used to detect the changes of LMP1 expression. The changes of MMP-9 were examined by RT-PCR assay and Western blot. The results were as follow: i) the colony formation inhibition rate (75.41 ± 3.9 percent in HNE1-LMP1 cells vs 37.89 ± 4.9 percent in HNE1 cells), the rate of attachment (HNE1-LMP1 vs HNE1: 56.40 ± 3.5 vs 65.87 ± 5.9 percent), the invasion inhibitory rate (HNE1-LMP1 vs HNE1: 56.50 ± 3.7 and 27.91 ± 2.1 percent), and the migration inhibitory rate (HNE1-LMP1 vs HNE1: 48.70 ± 3.9 vs 29.19 ± 6.27 percent) were all significantly different between the two cell lines (P < 0.01). ii) LMP1 was down-regulated in As2O3-treated HNE1-LMP1 cells. iii) The reduction of MMP-9 was found in As2O3-treated groups, more evident in HNE1-LMP1 cells. Thus, we conclude that As2O3 can reduce metastasis potential of NPC cells, involving inhibition of MMP-9 expression. LMP1 were also reduced in this process and seemed to enhance anti-metastasis activity of As2O3.


Assuntos
Humanos , Antineoplásicos/farmacologia , Arsenicais/farmacologia , Metaloproteinase 9 da Matriz/efeitos dos fármacos , Neoplasias Nasofaríngeas/tratamento farmacológico , Óxidos/farmacologia , Proteínas da Matriz Viral/efeitos dos fármacos , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Imunofluorescência , Regulação Neoplásica da Expressão Gênica , Microscopia Confocal , Metaloproteinase 9 da Matriz/genética , Neoplasias Nasofaríngeas/patologia , Invasividade Neoplásica/patologia , Metástase Neoplásica/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , RNA Mensageiro/efeitos dos fármacos , Proteínas da Matriz Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA