Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 121(21): e2401748121, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38739789

RESUMO

Potyviridae, the largest family of plant RNA viruses, includes many important pathogens that significantly reduce the yields of many crops worldwide. In this study, we report that the 6-kilodalton peptide 1 (6K1), one of the least characterized potyviral proteins, is an endoplasmic reticulum-localized protein. AI-assisted structure modeling and biochemical assays suggest that 6K1 forms pentamers with a central hydrophobic tunnel, can increase the cell membrane permeability of Escherichia coli and Nicotiana benthamiana, and can conduct potassium in Saccharomyces cerevisiae. An infectivity assay showed that viral proliferation is inhibited by mutations that affect 6K1 multimerization. Moreover, the 6K1 or its homologous 7K proteins from other viruses of the Potyviridae family also have the ability to increase cell membrane permeability and transmembrane potassium conductance. Taken together, these data reveal that 6K1 and its homologous 7K proteins function as viroporins in viral infected cells.


Assuntos
Nicotiana , Nicotiana/virologia , Nicotiana/metabolismo , Potyviridae/genética , Potyviridae/metabolismo , Proteínas Virais/metabolismo , Proteínas Virais/genética , Permeabilidade da Membrana Celular , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/virologia , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas Viroporinas/metabolismo , Proteínas Viroporinas/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Vírus de Plantas/genética , Vírus de Plantas/fisiologia , Doenças das Plantas/virologia , Potássio/metabolismo
2.
Curr Pharm Des ; 30(12): 912-920, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38482627

RESUMO

The discussion has revolved around the derivatives of amino acids and peptides containing carbocycles and their potential antiviral activity in vitro against influenza A, hepatitis C viruses, and coronavirus. Studies conducted on cell cultures reveal that aminoadamantane amino acid derivatives exhibit the capacity to hinder the replication of viruses containing viroporins. Furthermore, certain compounds demonstrate potent virucidal activity with respect to influenza A/H5N1 and hepatitis C virus particles. A conceptual framework for viroporin inhibitors has been introduced, incorporating carbocyclic motifs as membranotropic carriers in the structure, alongside a functional segment comprised of amino acids and peptides. These components correspond to the interaction with the inner surface of the channel's pore or another target protein.


Assuntos
Aminoácidos , Antivirais , Peptídeos , Antivirais/farmacologia , Antivirais/química , Antivirais/síntese química , Humanos , Peptídeos/farmacologia , Peptídeos/química , Peptídeos/síntese química , Aminoácidos/química , Aminoácidos/farmacologia , Animais , Hepacivirus/efeitos dos fármacos , Proteínas Viroporinas/antagonistas & inibidores , Proteínas Viroporinas/metabolismo , Proteínas Viroporinas/química , Vírus da Influenza A/efeitos dos fármacos
3.
PLoS Pathog ; 19(7): e1011217, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37494374

RESUMO

Many enveloped viruses enter host cells by fusing with acidic endosomes. The fusion activity of multiple viral envelope glycoproteins does not generally affect viral membrane permeability. However, fusion induced by the Lassa virus (LASV) glycoprotein complex (GPc) is always preceded by an increase in viral membrane permeability and the ensuing acidification of the virion interior. Here, systematic investigation of this LASV fusion phenotype using single pseudovirus tracking in live cells reveals that the change in membrane barrier function is associated with the fusogenic conformational reorganization of GPc. We show that a small-molecule fusion inhibitor or mutations that impair viral fusion by interfering with GPc refolding into the post-fusion structure prevent the increase in membrane permeability. We find that the increase in virion membrane permeability occurs early during endosomal maturation and is facilitated by virus-cell contact. This increase is observed using diverse arenavirus glycoproteins, whether presented on lentivirus-based pseudoviruses or arenavirus-like particles, and in multiple different cell types. Collectively, these results suggest that conformational changes in GPc triggered by low pH and cell factor binding are responsible for virion membrane permeabilization and acidification of the virion core prior to fusion. We propose that this viroporin-like activity may augment viral fusion and/or post-fusion steps of infection, including ribonucleoprotein release into the cytoplasm.


Assuntos
Arenavirus , Arenavirus/genética , Proteínas Viroporinas/metabolismo , Glicoproteínas/metabolismo , Proteínas do Envelope Viral/metabolismo , Vírus Lassa , Internalização do Vírus
4.
J Cell Sci ; 136(6)2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36807531

RESUMO

The coronavirus SARS-CoV-2, the agent of the deadly COVID-19 pandemic, is an enveloped virus propagating within the endocytic and secretory organelles of host mammalian cells. Enveloped viruses modify the ionic homeostasis of organelles to render their intra-luminal milieu permissive for viral entry, replication and egress. Here, we show that infection of Vero E6 cells with the delta variant of the SARS-CoV-2 alkalinizes the endoplasmic reticulum (ER)-Golgi intermediate compartment (ERGIC) as well as lysosomes, mimicking the effect of inhibitors of vacuolar proton ATPases. We further show the envelope protein of SARS-CoV-2 accumulates in the ERGIC when expressed in mammalian cells and selectively dissipates the ERGIC pH. This viroporin action is prevented by mutations of Val25 but not Asn15 within the channel pore of the envelope (E) protein. We conclude that the envelope protein acts as a proton channel in the ERGIC to mitigate the acidity of this intermediate compartment. The altered pH homeostasis of the ERGIC likely contributes to the virus fitness and pathogenicity, making the E channel an attractive drug target for the treatment of COVID-19.


Assuntos
COVID-19 , Proteínas do Envelope Viral , Animais , Humanos , Proteínas do Envelope Viral/metabolismo , Proteínas Viroporinas/metabolismo , COVID-19/metabolismo , Prótons , Pandemias , SARS-CoV-2/metabolismo , Complexo de Golgi/metabolismo , Lisossomos/metabolismo , Mamíferos/metabolismo
5.
J Virol ; 96(18): e0071622, 2022 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-36098511

RESUMO

Influenza A virus (IAV) assembly at the plasma membrane is orchestrated by at least five viral components, including hemagglutinin (HA), neuraminidase (NA), matrix (M1), the ion channel M2, and viral ribonucleoprotein (vRNP) complexes, although particle formation is observed with expression of only HA and/or NA. While these five viral components are expressed efficiently in primary human monocyte-derived macrophages (MDMs) upon IAV infection, this cell type does not support efficient HA-M2 association and IAV particle assembly at the plasma membrane. Both defects are specific to MDMs and can be reversed upon disruption of F-actin. However, the relationship between the two defects is unclear. Here, we examined whether M2 contributes to particle assembly in MDMs and if so, which region of M2 determines the susceptibility to the MDM-specific and actin-dependent suppression. An analysis using correlative fluorescence and scanning electron microscopy showed that an M2-deficient virus failed to form budding structures at the cell surface even after F-actin was disrupted, indicating that M2 is essential for virus particle formation at the MDM surface. Notably, proximity ligation analysis revealed that a single amino acid substitution in a Glu-Glu-Tyr sequence (residues 74 to 76) in the M2 cytoplasmic tail allowed the HA-M2 association to occur efficiently even in MDMs with intact actin cytoskeleton. This phenotype did not correlate with known phenotypes of the M2 substitution mutants regarding M1 interaction or vRNP packaging in epithelial cells. Overall, our study identified M2 as a target of MDM-specific restriction of IAV assembly, which requires the Glu-Glu-Tyr sequence in the cytoplasmic tail. IMPORTANCE Human MDMs represent a cell type that is nonpermissive to particle formation of influenza A virus (IAV). We previously showed that close proximity association between viral HA and M2 proteins is blocked in MDMs. However, whether MDMs express a restriction factor against IAV assembly or whether they lack a dependency factor promoting assembly remained unknown. In the current study, we determined that the M2 protein is necessary for particle formation in MDMs but is also a molecular target of the MDM-specific suppression of assembly. Substitutions in the M2 cytoplasmic tail alleviated the block in both the HA-M2 association and particle production in MDMs. These findings suggest that MDMs express dependency factors necessary for assembly but also express a factor(s) that inhibits HA-M2 association and particle formation. High conservation of the M2 sequence rendering the susceptibility to the assembly block highlights the potential for M2 as a target of antiviral strategies.


Assuntos
Ácido Glutâmico , Hemaglutininas , Vírus da Influenza A , Macrófagos , Tirosina , Proteínas da Matriz Viral , Proteínas Viroporinas , Montagem de Vírus , Actinas/metabolismo , Sequência de Aminoácidos , Ácido Glutâmico/genética , Hemaglutininas/metabolismo , Interações entre Hospedeiro e Microrganismos/genética , Humanos , Vírus da Influenza A/genética , Vírus da Influenza A/metabolismo , Macrófagos/virologia , Neuraminidase/genética , Neuraminidase/metabolismo , Ribonucleoproteínas/genética , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/metabolismo , Proteínas Viroporinas/química , Proteínas Viroporinas/metabolismo , Montagem de Vírus/genética
6.
PLoS Pathog ; 18(2): e1010310, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35130329

RESUMO

Recent studies identified signal peptidase complex subunit 1 (SPCS1) as a proviral host factor for Flaviviridae viruses, including HCV. One of the SPCS1's roles in flavivirus propagation was attributed to its regulation of signal peptidase complex (SPC)-mediated processing of flavivirus polyprotein, especially C-prM junction. However, whether SPCS1 also regulates any SPC-mediated processing sites within HCV polyprotein remains unclear. In this study, we determined that loss of SPCS1 specifically impairs the HCV E2-p7 processing by the SPC. We also determined that efficient separation of E2 and p7, regardless of its dependence on SPC-mediated processing, leads to SPCS1 dispensable for HCV assembly These results suggest that SPCS1 regulates HCV assembly by facilitating the SPC-mediated processing of E2-p7 precursor. Structural modeling suggests that intrinsically delayed processing of the E2-p7 is likely caused by the structural rigidity of p7 N-terminal transmembrane helix-1 (p7/TM1/helix-1), which has mostly maintained membrane-embedded conformations during molecular dynamics (MD) simulations. E2-p7-processing-impairing p7 mutations narrowed the p7/TM1/helix-1 bending angle against the membrane, resulting in closer membrane embedment of the p7/TM1/helix-1 and less access of E2-p7 junction substrate to the catalytic site of the SPC, located well above the membrane in the ER lumen. Based on these results we propose that the key mechanism of action of SPCS1 in HCV assembly is to facilitate the E2-p7 processing by enhancing the E2-p7 junction site presentation to the SPC active site. By providing evidence that SPCS1 facilitates HCV assembly by regulating SPC-mediated cleavage of E2-p7 junction, equivalent to the previously established role of this protein in C-prM junction processing in flavivirus, this study establishes the common role of SPCS1 in Flaviviridae family virus propagation as to exquisitely regulate the SPC-mediated processing of specific, suboptimal target sites.


Assuntos
Hepacivirus/metabolismo , Hepatite C/virologia , Proteínas de Membrana/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas Viroporinas/metabolismo , Montagem de Vírus , Linhagem Celular , Células HEK293 , Interações entre Hospedeiro e Microrganismos , Humanos , Proteínas de Membrana/química , Simulação de Dinâmica Molecular , Conformação Proteica , Proteínas do Envelope Viral/química , Proteínas Viroporinas/química , Replicação Viral
7.
Sci Rep ; 12(1): 1005, 2022 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-35046461

RESUMO

The pandemic of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has caused a public health emergency, and research on the development of various types of vaccines is rapidly progressing at an unprecedented development speed internationally. Some vaccines have already been approved for emergency use and are being supplied to people around the world, but there are still many ongoing efforts to create new vaccines. Virus-like particles (VLPs) enable the construction of promising platforms in the field of vaccine development. Here, we demonstrate that non-infectious SARS-CoV-2 VLPs can be successfully assembled by co-expressing three important viral proteins membrane (M), envelop (E) and nucleocapsid (N) in plants. Plant-derived VLPs were purified by sedimentation through a sucrose cushion. The shape and size of plant-derived VLPs are similar to native SARS-CoV-2 VLPs without spike. Although the assembled VLPs do not have S protein spikes, they could be developed as formulations that can improve the immunogenicity of vaccines including S antigens, and further could be used as platforms that can carry S antigens of concern for various mutations.


Assuntos
Vacinas contra COVID-19/imunologia , COVID-19/imunologia , Proteínas M de Coronavírus/imunologia , Proteínas do Nucleocapsídeo de Coronavírus/imunologia , SARS-CoV-2/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Proteínas Viroporinas/imunologia , COVID-19/prevenção & controle , COVID-19/virologia , Vacinas contra COVID-19/administração & dosagem , Proteínas M de Coronavírus/genética , Proteínas M de Coronavírus/metabolismo , Proteínas do Nucleocapsídeo de Coronavírus/genética , Proteínas do Nucleocapsídeo de Coronavírus/metabolismo , Humanos , Nicotiana/imunologia , Nicotiana/metabolismo , Nicotiana/virologia , Vacinas de Partículas Semelhantes a Vírus/genética , Vacinas de Partículas Semelhantes a Vírus/metabolismo , Proteínas Viroporinas/genética , Proteínas Viroporinas/metabolismo
8.
Viruses ; 13(11)2021 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-34834972

RESUMO

The current COVID-19 pandemic has highlighted the need for the research community to develop a better understanding of viruses, in particular their modes of infection and replicative lifecycles, to aid in the development of novel vaccines and much needed anti-viral therapeutics. Several viruses express proteins capable of forming pores in host cellular membranes, termed "Viroporins". They are a family of small hydrophobic proteins, with at least one amphipathic domain, which characteristically form oligomeric structures with central hydrophilic domains. Consequently, they can facilitate the transport of ions through the hydrophilic core. Viroporins localise to host membranes such as the endoplasmic reticulum and regulate ion homeostasis creating a favourable environment for viral infection. Viroporins also contribute to viral immune evasion via several mechanisms. Given that viroporins are often essential for virion assembly and egress, and as their structural features tend to be evolutionarily conserved, they are attractive targets for anti-viral therapeutics. This review discusses the current knowledge of several viroporins, namely Influenza A virus (IAV) M2, Human Immunodeficiency Virus (HIV)-1 Viral protein U (Vpu), Hepatitis C Virus (HCV) p7, Human Papillomavirus (HPV)-16 E5, Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) Open Reading Frame (ORF)3a and Polyomavirus agnoprotein. We highlight the intricate but broad immunomodulatory effects of these viroporins and discuss the current antiviral therapies that target them; continually highlighting the need for future investigations to focus on novel therapeutics in the treatment of existing and future emergent viruses.


Assuntos
Imunomodulação , Canais Iônicos/metabolismo , Proteínas Viroporinas/metabolismo , Viroses/tratamento farmacológico , Vírus/metabolismo , Antivirais/farmacologia , Antivirais/uso terapêutico , Autofagia , Interações Hospedeiro-Patógeno , Proteínas do Vírus da Imunodeficiência Humana/química , Proteínas do Vírus da Imunodeficiência Humana/metabolismo , Evasão da Resposta Imune , Inflamassomos/imunologia , Proteínas Oncogênicas Virais/química , Proteínas Oncogênicas Virais/metabolismo , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/metabolismo , Proteínas Virais/química , Proteínas Virais/metabolismo , Proteínas Virais Reguladoras e Acessórias/química , Proteínas Virais Reguladoras e Acessórias/metabolismo , Proteínas Estruturais Virais/química , Proteínas Estruturais Virais/metabolismo , Proteínas Viroporinas/química , Viroses/imunologia , Viroses/virologia , Vírus/efeitos dos fármacos , Vírus/imunologia , Vírus/patogenicidade
9.
Viruses ; 13(10)2021 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-34696358

RESUMO

Recently, two cases of complete remission of classical Hodgkin lymphoma (cHL) and follicular lymphoma (FL) after SARS-CoV-2 infection were reported. However, the precise molecular mechanism of this rare event is yet to be understood. Here, we hypothesize a potential anti-tumor immune response of SARS-CoV-2 and based on a computational approach show that: (i) SARS-CoV-2 Spike-RBD may bind to the extracellular domains of CD15, CD27, CD45, and CD152 receptors of cHL or FL and may directly inhibit cell proliferation. (ii) Alternately, upon internalization after binding to these CD molecules, the SARS-CoV-2 membrane (M) protein and ORF3a may bind to gamma-tubulin complex component 3 (GCP3) at its tubulin gamma-1 chain (TUBG1) binding site. (iii) The M protein may also interact with TUBG1, blocking its binding to GCP3. (iv) Both the M and ORF3a proteins may render the GCP2-GCP3 lateral binding where the M protein possibly interacts with GCP2 at its GCP3 binding site and the ORF3a protein to GCP3 at its GCP2 interacting residues. (v) Interactions of the M and ORF3a proteins with these gamma-tubulin ring complex components potentially block the initial process of microtubule nucleation, leading to cell-cycle arrest and apoptosis. (vi) The Spike-RBD may also interact with and block PD-1 signaling similar to pembrolizumab and nivolumab- like monoclonal antibodies and may induce B-cell apoptosis and remission. (vii) Finally, the TRADD interacting "PVQLSY" motif of Epstein-Barr virus LMP-1, that is responsible for NF-kB mediated oncogenesis, potentially interacts with SARS-CoV-2 Mpro, NSP7, NSP10, and spike (S) proteins, and may inhibit the LMP-1 mediated cell proliferation. Taken together, our results suggest a possible therapeutic potential of SARS-CoV-2 in lymphoproliferative disorders.


Assuntos
COVID-19/metabolismo , Linfoma/imunologia , SARS-CoV-2/imunologia , Anticorpos Monoclonais/imunologia , Antineoplásicos/farmacologia , Sítios de Ligação , COVID-19/complicações , Glicoproteínas/metabolismo , Glicoproteínas/ultraestrutura , Humanos , Imunidade/imunologia , Linfoma/terapia , Linfoma/virologia , Modelos Teóricos , Simulação de Acoplamento Molecular , Ligação Proteica , Domínios Proteicos , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/ultraestrutura , Proteínas Viroporinas/metabolismo , Proteínas Viroporinas/ultraestrutura
10.
Mediators Inflamm ; 2021: 1267041, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34483726

RESUMO

HIV-1 can incite activation of chemokine receptors, inflammatory mediators, and glutamate receptor-mediated excitotoxicity. The mechanisms associated with such immune activation can disrupt neuronal and glial functions. HIV-associated neurocognitive disorder (HAND) is being observed since the beginning of the AIDS epidemic due to a change in the functional integrity of cells from the central nervous system (CNS). Even with the presence of antiretroviral therapy, there is a decline in the functioning of the brain especially movement skills, noticeable swings in mood, and routine performance activities. Under the umbrella of HAND, various symptomatic and asymptomatic conditions are categorized and are on a rise despite the use of newer antiretroviral agents. Due to the use of long-lasting antiretroviral agents, this deadly disease is becoming a manageable chronic condition with the occurrence of asymptomatic neurocognitive impairment (ANI), symptomatic mild neurocognitive disorder, or HIV-associated dementia. In-depth research in the pathogenesis of HIV has focused on various mechanisms involved in neuronal dysfunction and associated toxicities ultimately showcasing the involvement of various pathways. Increasing evidence-based studies have emphasized a need to focus and explore the specific pathways in inflammation-associated neurodegenerative disorders. In the current review, we have highlighted the association of various HIV proteins and neuronal cells with their involvement in various pathways responsible for the development of neurotoxicity.


Assuntos
Complexo AIDS Demência/imunologia , Complexo AIDS Demência/virologia , Sistema Nervoso Central/virologia , HIV-1/metabolismo , Proteínas Virais/metabolismo , Complexo AIDS Demência/fisiopatologia , Antirretrovirais/uso terapêutico , Astrócitos/virologia , Sistema Nervoso Central/fisiopatologia , Genoma , Proteína gp120 do Envelope de HIV/metabolismo , Proteína gp41 do Envelope de HIV/metabolismo , Infecções por HIV/complicações , Infecções por HIV/metabolismo , Proteínas do Vírus da Imunodeficiência Humana/metabolismo , Humanos , Inflamação , Cinurenina/metabolismo , Macrófagos/virologia , Microglia/virologia , Neurônios/virologia , Oligodendroglia/virologia , Receptores de N-Metil-D-Aspartato/metabolismo , Carga Viral , Proteínas Virais Reguladoras e Acessórias/metabolismo , Proteínas Viroporinas/metabolismo , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo , Produtos do Gene rev do Vírus da Imunodeficiência Humana/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo
11.
mBio ; 12(4): e0192021, 2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34425695

RESUMO

Human immunodeficiency virus (HIV) remodels the cell surface of infected cells to facilitate viral dissemination and promote immune evasion. The membrane-associated viral protein U (Vpu) accessory protein encoded by HIV-1 plays a key role in this process by altering cell surface levels of multiple host proteins. Using an unbiased quantitative plasma membrane profiling approach, we previously identified CD47 as a putative host target downregulated by Vpu. CD47 is a ubiquitously expressed cell surface protein that interacts with the myeloid cell inhibitory receptor signal regulatory protein-alpha (SIRPα) to deliver a "don't-eat-me" signal, thus protecting cells from phagocytosis. In this study, we investigate whether CD47 modulation by HIV-1 Vpu might promote the susceptibility of macrophages to viral infection via phagocytosis of infected CD4+ T cells. Indeed, we find that Vpu downregulates CD47 expression on infected CD4+ T cells, leading to enhanced capture and phagocytosis by macrophages. We further provide evidence that this Vpu-dependent process allows a C-C chemokine receptor type 5 (CCR5)-tropic transmitted/founder (T/F) virus, which otherwise poorly infects macrophages in its cell-free form, to efficiently infect macrophages. Importantly, we show that HIV-1-infected cells expressing a Vpu-resistant CD47 mutant are less prone to infecting macrophages through phagocytosis. Mechanistically, Vpu forms a physical complex with CD47 through its transmembrane domain and targets the latter for lysosomal degradation. These results reveal a novel role of Vpu in modulating macrophage infection, which has important implications for HIV-1 transmission in early stages of infection and the establishment of viral reservoir. IMPORTANCE Macrophages play critical roles in human immunodeficiency virus (HIV) transmission, viral spread early in infection, and as a reservoir of virus. Selective capture and engulfment of HIV-1-infected T cells was shown to drive efficient macrophage infection, suggesting that this mechanism represents an important mode of infection notably for weakly macrophage-tropic T/F viruses. In this study, we provide insight into the signals that regulate this process. We show that the HIV-1 accessory protein viral protein U (Vpu) downregulates cell surface levels of CD47, a host protein that interacts with the inhibitory receptor signal regulatory protein-alpha (SIRPα), to deliver a "don't-eat-me" signal to macrophages. This allows for enhanced capture and phagocytosis of infected T cells by macrophages, ultimately leading to their productive infection even with transmitted/founder (T/F) virus. These findings provide new insights into the mechanisms governing the intercellular transmission of HIV-1 to macrophages with implications for the establishment of the macrophage reservoir and early HIV-1 dissemination in vivo.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Antígeno CD47/genética , Regulação para Baixo , HIV-1/química , HIV-1/imunologia , Proteínas do Vírus da Imunodeficiência Humana/genética , Macrófagos/virologia , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Viroporinas/genética , Linfócitos T CD4-Positivos/virologia , Antígeno CD47/imunologia , Células HEK293 , Proteínas do Vírus da Imunodeficiência Humana/metabolismo , Humanos , Células Jurkat , Macrófagos/imunologia , Fagocitose , Proteínas Virais Reguladoras e Acessórias/metabolismo , Proteínas Viroporinas/metabolismo
12.
Viruses ; 13(3)2021 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-33807095

RESUMO

The etiological agent of the COVID-19 pandemic is SARS-CoV-2. As a member of the Coronaviridae, the enveloped pathogen has several membrane proteins, of which two, E and 3a, were suggested to function as ion channels. In an effort to increase our treatment options, alongside providing new research tools, we have sought to inhibit the 3a channel by targeted drug repurposing. To that end, using three bacteria-based assays, we screened a library of 2839 approved-for-human-use drugs and identified the following potential channel-blockers: Capreomycin, Pentamidine, Spectinomycin, Kasugamycin, Plerixafor, Flumatinib, Litronesib, Darapladib, Floxuridine and Fludarabine. The stage is now set for examining the activity of these compounds in detailed electrophysiological studies and their impact on the whole virus with appropriate biosafety measures.


Assuntos
Antivirais/farmacologia , COVID-19/virologia , Reposicionamento de Medicamentos , SARS-CoV-2/efeitos dos fármacos , Proteínas do Envelope Viral/antagonistas & inibidores , Proteínas do Envelope Viral/metabolismo , Proteínas Viroporinas/antagonistas & inibidores , Proteínas Viroporinas/metabolismo , Avaliação Pré-Clínica de Medicamentos , Humanos , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Proteínas do Envelope Viral/genética , Proteínas Viroporinas/genética , Tratamento Farmacológico da COVID-19
13.
Virology ; 553: 135-153, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33278736

RESUMO

JC virus encodes an important regulatory protein, known as Agnoprotein (Agno). We have recently reported Agno's first protein-interactome with its cellular partners revealing that it targets various cellular networks and organelles, including mitochondria. Here, we report further characterization of the functional consequences of its mitochondrial targeting and demonstrated its co-localization with the mitochondrial networks and with the mitochondrial outer membrane. The mitochondrial targeting sequence (MTS) of Agno and its dimerization domain together play major roles in this targeting. Data also showed alterations in various mitochondrial functions in Agno-positive cells; including a significant reduction in mitochondrial membrane potential, respiration rates and ATP production. In contrast, a substantial increase in ROS production and Ca2+ uptake by the mitochondria were also observed. Finally, findings also revealed a significant decrease in viral replication when Agno MTS was deleted, highlighting a role for MTS in the function of Agno during the viral life cycle.


Assuntos
Vírus JC/metabolismo , Mitocôndrias/metabolismo , Proteínas Virais Reguladoras e Acessórias/metabolismo , Proteínas Viroporinas/metabolismo , Trifosfato de Adenosina/metabolismo , Cálcio/metabolismo , Respiração Celular , Dimerização , Humanos , Potencial da Membrana Mitocondrial , Membranas Mitocondriais/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas Virais Reguladoras e Acessórias/química , Proteínas Viroporinas/química , Replicação Viral
14.
Front Immunol ; 11: 573339, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33154751

RESUMO

Coronavirus (CoV) outbreaks have recently emerged as a global public health threat due to their exceptional zoonotic potential - a feature arising from their ability to infect a diverse range of potential hosts combined with their high capacity for mutation and recombination. After Severe Acute Respiratory Syndrome (SARS) CoV-1 in 2003 and Middle East Respiratory Syndrome (MERS) CoV in 2012, with the current SARS-CoV-2 pandemic we are now in the midst of the third deadly international CoV outbreak in less than 20 years. Coronavirus outbreaks present a critical threat to global public health and an urgent necessity for therapeutic options. Here, we critically examine the current evidence for ion channel activity in CoV proteins and the potential for modulation as a therapeutic approach.


Assuntos
Canais Iônicos/metabolismo , Síndrome Respiratória Aguda Grave/virologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/metabolismo , Proteínas Virais/metabolismo , Proteínas Viroporinas/metabolismo , Animais , Humanos , Canais Iônicos/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/genética , Proteínas Viroporinas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA