Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Intervalo de año de publicación
1.
Cell Rep ; 42(11): 113285, 2023 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-37910505

RESUMEN

Deciphering the mechanisms underlying viral persistence is critical to achieving a cure for human immunodeficiency virus (HIV) infection. Here, we implement a systems approach to discover molecular signatures of HIV latently infected CD4+ T cells, identifying the immunosuppressive, adenosine-producing ectonucleotidase CD73 as a key surface marker of latent cells. Hypoxic conditioning, reflecting the lymphoid tissue microenvironment, increases the frequency of CD73+ CD4+ T cells and promotes HIV latency. Transcriptomic profiles of CD73+ CD4+ T cells favor viral quiescence, immune evasion, and cell survival. CD73+ CD4+ T cells are capable of harboring a functional HIV reservoir and reinitiating productive infection ex vivo. CD73 or adenosine receptor blockade facilitates latent HIV reactivation in vitro, mechanistically linking adenosine signaling to viral quiescence. Finally, tissue imaging of lymph nodes from HIV-infected individuals on antiretroviral therapy reveals spatial association between CD73 expression and HIV persistence in vivo. Our findings warrant development of HIV-cure strategies targeting the hypoxia-CD73-adenosine axis.


Asunto(s)
Infecciones por VIH , VIH-1 , Humanos , Adenosina/metabolismo , Linfocitos T CD4-Positivos , Activación Viral , Latencia del Virus/fisiología , Replicación Viral/fisiología
2.
J Virol ; 96(4): e0162221, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-34935434

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can induce mild to life-threatening symptoms. Especially individuals over 60 years of age or with underlying comorbidities, including heart or lung disease and diabetes, or immunocompromised patients are at a higher risk. Fatal multiorgan damage in coronavirus disease 2019 (COVID-19) patients can be attributed to an interleukin-6 (IL-6)-dominated cytokine storm. Consequently, IL-6 receptor (IL-6R) monoclonal antibody treatment for severe COVID-19 cases has been approved for therapy. High concentrations of soluble IL-6R (sIL-6R) were found in COVID-19 intensive care unit patients, suggesting the involvement of IL-6 trans-signaling in disease pathology. Here, in analogy to bispecific antibodies (bsAbs), we developed the first bispecific IL-6 trans-signaling inhibitor, c19s130Fc, which blocks viral infection and IL-6 trans-signaling. c19s130Fc is a designer protein of the IL-6 trans-signaling inhibitor cs130 fused to a single-domain nanobody directed against the receptor binding domain (RBD) of the SARS-CoV-2 spike protein. c19s130Fc binds with high affinity to IL-6:sIL-6R complexes as well as the spike protein of SARS-CoV-2, as shown by surface plasmon resonance. Using cell-based assays, we demonstrate that c19s130Fc blocks IL-6 trans-signaling-induced proliferation and STAT3 phosphorylation in Ba/F3-gp130 cells as well as SARS-CoV-2 infection and STAT3 phosphorylation in Vero cells. Taken together, c19s130Fc represents a new class of bispecific inhibitors consisting of a soluble cytokine receptor fused to antiviral nanobodies and principally demonstrates the multifunctionalization of trans-signaling inhibitors. IMPORTANCE The availability of effective SARS-CoV-2 vaccines is a large step forward in managing the pandemic situation. In addition, therapeutic options, e.g., monoclonal antibodies to prevent viral cell entry and anti-inflammatory therapies, including glucocorticoid treatment, are currently developed or in clinical use to treat already infected patients. Here, we report a novel dual-specificity inhibitor to simultaneously target SARS-CoV-2 infection and virus-induced hyperinflammation. This was achieved by fusing an inhibitor of viral cell entry with a molecule blocking IL-6, a key mediator of SARS-CoV-2-induced hyperinflammation. Through this dual action, this molecule may have the potential to efficiently ameliorate symptoms of COVID-19 in infected individuals.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19 , Receptor gp130 de Citocinas , Interleucina-6/metabolismo , Proteínas Recombinantes de Fusión , Transducción de Señal/efectos de los fármacos , Anticuerpos de Dominio Único , Glicoproteína de la Espiga del Coronavirus/metabolismo , Animales , COVID-19/metabolismo , Chlorocebus aethiops , Receptor gp130 de Citocinas/química , Receptor gp130 de Citocinas/genética , Humanos , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/farmacología , Células Vero
3.
Cell ; 179(4): 880-894.e10, 2019 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-31668804

RESUMEN

Current approaches to reducing the latent HIV reservoir entail first reactivating virus-containing cells to become visible to the immune system. A critical second step is killing these cells to reduce reservoir size. Endogenous cytotoxic T-lymphocytes (CTLs) may not be adequate because of cellular exhaustion and the evolution of CTL-resistant viruses. We have designed a universal CAR-T cell platform based on CTLs engineered to bind a variety of broadly neutralizing anti-HIV antibodies. We show that this platform, convertibleCAR-T cells, effectively kills HIV-infected, but not uninfected, CD4 T cells from blood, tonsil, or spleen and only when armed with anti-HIV antibodies. convertibleCAR-T cells also kill within 48 h more than half of the inducible reservoir found in blood of HIV-infected individuals on antiretroviral therapy. The modularity of convertibleCAR-T cell system, which allows multiplexing with several anti-HIV antibodies yielding greater breadth and control, makes it a promising tool for attacking the latent HIV reservoir.


Asunto(s)
Anticuerpos Antiidiotipos/farmacología , Infecciones por VIH/terapia , Inmunoterapia Adoptiva , Replicación Viral/genética , Animales , Anticuerpos Antiidiotipos/inmunología , Células HEK293 , Infecciones por VIH/genética , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/inmunología , VIH-1/patogenicidad , Humanos , Ratones , Tonsila Palatina/inmunología , Tonsila Palatina/metabolismo , Cultivo Primario de Células , Bazo/inmunología , Bazo/metabolismo , Linfocitos T Citotóxicos/inmunología , Latencia del Virus/inmunología , Replicación Viral/inmunología
4.
Sci Rep ; 9(1): 707, 2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30679542

RESUMEN

Hantavirus assembly and budding are governed by the surface glycoproteins Gn and Gc. In this study, we investigated the glycoproteins of Puumala, the most abundant Hantavirus species in Europe, using fluorescently labeled wild-type constructs and cytoplasmic tail (CT) mutants. We analyzed their intracellular distribution, co-localization and oligomerization, applying comprehensive live, single-cell fluorescence techniques, including confocal microscopy, imaging flow cytometry, anisotropy imaging and Number&Brightness analysis. We demonstrate that Gc is significantly enriched in the Golgi apparatus in absence of other viral components, while Gn is mainly restricted to the endoplasmic reticulum (ER). Importantly, upon co-expression both glycoproteins were found in the Golgi apparatus. Furthermore, we show that an intact CT of Gc is necessary for efficient Golgi localization, while the CT of Gn influences protein stability. Finally, we found that Gn assembles into higher-order homo-oligomers, mainly dimers and tetramers, in the ER while Gc was present as mixture of monomers and dimers within the Golgi apparatus. Our findings suggest that PUUV Gc is the driving factor of the targeting of Gc and Gn to the Golgi region, while Gn possesses a significantly stronger self-association potential.


Asunto(s)
Glicoproteínas/metabolismo , Infecciones por Hantavirus/metabolismo , Fiebre Hemorrágica con Síndrome Renal/metabolismo , Multimerización de Proteína , Virus Puumala/fisiología , Fracciones Subcelulares/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Membrana Celular/metabolismo , Membrana Celular/virología , Chlorocebus aethiops , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Glicoproteínas/química , Aparato de Golgi/metabolismo , Aparato de Golgi/virología , Células HEK293 , Infecciones por Hantavirus/virología , Fiebre Hemorrágica con Síndrome Renal/virología , Humanos , Proteínas Luminiscentes/metabolismo , Fracciones Subcelulares/virología , Células Vero , Proteínas del Envoltorio Viral/química
5.
Biochemistry ; 58(6): 818-832, 2019 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-30602116

RESUMEN

The human immunodeficiency virus enters its host cells by membrane fusion, initiated by the gp41 subunit of its envelope protein. gp41 has also been shown to bind T-cell receptor (TCR) complex components, interfering with TCR signaling leading to reduced T-cell activation. This immunoinhibitory activity is suggested to occur during the membrane fusion process and is attributed to various membranotropic regions of the gp41 ectodomain and to the transmembrane domain. Although extensively studied, the cytosolic region of gp41, termed the cytoplasmic tail (CT), has not been examined in the context of immune suppression. Here we investigated whether the CT inhibits T-cell activation in different T-cell models by utilizing gp41-derived peptides and expressed full gp41 proteins. We found that a conserved region of the CT, termed lentiviral lytic peptide 2 (LLP2), specifically inhibits the activation of mouse, Jurkat, and human primary T-cells. This inhibition resulted in reduced T-cell proliferation, gene expression, cytokine secretion, and cell surface expression of CD69. Differential activation of the TCR signaling cascade revealed that CT-based immune suppression occurs downstream of the TCR complex. Moreover, LLP2 peptide treatment of Jurkat and primary human T-cells impaired Akt but not NFκB and ERK1/2 activation, suggesting that immune suppression occurs through the Akt pathway. These findings identify a novel gp41 T-cell suppressive element with a unique inhibitory mechanism that can take place post-membrane fusion.


Asunto(s)
Proteína gp41 de Envoltorio del VIH/inmunología , VIH-1/inmunología , Activación de Linfocitos/inmunología , Linfocitos T/inmunología , Secuencias de Aminoácidos , Animales , Proliferación Celular , Citocinas/genética , Citocinas/metabolismo , Expresión Génica , Proteína gp41 de Envoltorio del VIH/química , Humanos , Células Jurkat , Ratones , Ratones Endogámicos C57BL , Fosforilación , Dominios Proteicos , Proteínas Proto-Oncogénicas c-akt/química , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células RAW 264.7 , Transducción de Señal , Linfocitos T/metabolismo , Linfocitos T/virología , Serina-Treonina Quinasas TOR/química , Serina-Treonina Quinasas TOR/metabolismo
6.
Cell Host Microbe ; 21(5): 569-579.e6, 2017 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-28494238

RESUMEN

Transcriptional latency of HIV is a last barrier to viral eradication. Chromatin-remodeling complexes and post-translational histone modifications likely play key roles in HIV-1 reactivation, but the underlying mechanisms are incompletely understood. We performed an RNAi-based screen of human lysine methyltransferases and identified the SET and MYND domain-containing protein 2 (SMYD2) as an enzyme that regulates HIV-1 latency. Knockdown of SMYD2 or its pharmacological inhibition reactivated latent HIV-1 in T cell lines and in primary CD4+ T cells. SMYD2 associated with latent HIV-1 promoter chromatin, which was enriched in monomethylated lysine 20 at histone H4 (H4K20me1), a mark lost in cells lacking SMYD2. Further, we find that lethal 3 malignant brain tumor 1 (L3MBTL1), a reader protein with chromatin-compacting properties that recognizes H4K20me1, was recruited to the latent HIV-1 promoter in a SMYD2-dependent manner. We propose that a SMYD2-H4K20me1-L3MBTL1 axis contributes to HIV-1 latency and can be targeted with small-molecule SMYD2 inhibitors.


Asunto(s)
VIH-1/fisiología , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Latencia del Virus/fisiología , Linfocitos T CD4-Positivos , Línea Celular , Cromatina/química , Cromatina/genética , Proteínas Cromosómicas no Histona/metabolismo , ADN Recombinante , Femenino , Células HEK293 , VIH-1/genética , N-Metiltransferasa de Histona-Lisina/genética , Histonas/química , Humanos , Lisina/metabolismo , Metilación , Regiones Promotoras Genéticas , ARN Interferente Pequeño , Proteínas Represoras , Linfocitos T/virología , Proteínas Supresoras de Tumor
7.
Cell Microbiol ; 18(1): 125-36, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26243691

RESUMEN

Viral glycoproteins are highly variable in their primary structure, but on the other hand feature a high functional conservation to fulfil their versatile tasks during the pathogenic life cycle. Typically, all protein domains are optimized in that indispensable functions can be assigned to small conserved motifs or even individual amino acids. The cytoplasmic tail of many viral spike proteins, although of particular relevance for the virus biology, is often only insufficiently characterized. Hemagglutinin (HA), the receptor-binding protein of the influenza virus comprises a short cytoplasmic tail of 13 amino acids that exhibits three highly conserved palmitoylation sites. However, the particular importance of these modifications and the tail in general for intracellular trafficking and lateral membrane organization remains elusive. In this study, we generated HA core proteins consisting of transmembrane domain, cytoplasmic tail and a minor part of the ectodomain, tagged with a yellow fluorescent protein. Different mutation and truncation variants of these chimeric proteins were investigated using confocal microscopy, to characterize the role of cytoplasmic tail and palmitoylation for the intracellular trafficking to plasma membrane and Golgi apparatus. In addition, we assessed raft partitioning of the variants by Foerster resonance energy transfer with an established raft marker. We revealed a substantial influence of the cytoplasmic tail length on the intracellular distribution and surface exposure of the proteins. A complete removal of the tail hampers a physiological trafficking of the protein, whereas a partial truncation can be compensated by cytoplasmic palmitoylations. Plasma membrane raft partitioning on the other hand was found to imperatively require palmitoylations, and the cysteine at position 551 turned out to be of most relevance. Our data shed further light on the tight interconnection between cytoplasmic elements and intracellular trafficking and suggest a function of HA palmitoylations in both lateral sorting and anterograde trafficking of the glycoprotein.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Interacciones Huésped-Patógeno , Microdominios de Membrana/metabolismo , Orthomyxoviridae/fisiología , Animales , Proteínas Bacterianas/análisis , Proteínas Bacterianas/genética , Células CHO , Cricetulus , Aparato de Golgi/metabolismo , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/genética , Microscopía Confocal , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Proteínas Recombinantes de Fusión/análisis , Proteínas Recombinantes de Fusión/genética
8.
Chembiochem ; 16(9): 1288-92, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25882139

RESUMEN

We have established a method of preparing giant plasma membrane vesicles (GPMVs) by using cysteine mutants of the proapoptotic peptide (PAP) Ac-R7-GG-KLAKLAKKLAKLAK. A cysteine scan revealed that cytotoxicity and GPMV formation were dependent on the cysteine position within the PAP sequence. In comparison to GPMVs prepared by extensive treatment with paraformaldehyde (PFA) and dithiothreitol (DTT), our GPMVs were produced from HeLa cells at much lower concentrations of the blebbing agent. We found that only GPMVs derived from cysteine-containing PAP showed lipid phase separation. This membrane model was applied to investigate the phase partitioning of two relevant membrane proteins: influenza virus hemagglutinin (HA) and tetherin, which clamps budding HIV to infected cells. For tetherin, we show for the first time exclusion from cholesterol-rich domains in a GPMV model, thus documenting the potential of our approach for membrane-partitioning studies.


Asunto(s)
Membrana Celular/metabolismo , Cisteína/metabolismo , Microdominios de Membrana/metabolismo , Péptidos/metabolismo , Secuencia de Aminoácidos , Antígenos CD/metabolismo , Colesterol/metabolismo , Cisteína/química , Proteínas Ligadas a GPI/metabolismo , Células HeLa , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Humanos , Datos de Secuencia Molecular , Orthomyxoviridae/fisiología , Péptidos/química , Transición de Fase
9.
Angew Chem Int Ed Engl ; 54(1): 323-6, 2015 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-25417776

RESUMEN

This study presents a novel and easily applicable approach to recruit sulfhydryl-containing biomolecules to membranes by using a palmitic acid which is functionalized with a maleimide group. Notably, this strategy can also be employed with preformed (biological) membranes. The applicability of the assay is demonstrated by characterizing the binding of a Rhodamine-labeled peptide to lipid and cellular membranes using methods of fluorescence spectroscopy, lifetime measurement, and microscopy. Our approach offers new possibilities for preparing biologically active liposomes and manipulating living cells.


Asunto(s)
Membrana Celular/metabolismo , Liposomas/metabolismo , Maleimidas/metabolismo , Ácido Palmítico/metabolismo , Péptidos/metabolismo , Compuestos de Sulfhidrilo/metabolismo , Liposomas/química , Macrófagos/citología , Macrófagos/metabolismo , Maleimidas/química , Microscopía Confocal , Ácido Palmítico/química , Péptidos/química , Espectrometría de Fluorescencia , Compuestos de Sulfhidrilo/análisis
10.
PLoS Pathog ; 10(8): e1004248, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25121610

RESUMEN

HIV-1 uses a number of means to manipulate the immune system, to avoid recognition and to highjack signaling pathways. HIV-1 infected cells show limited Toll-Like Receptor (TLR) responsiveness via as yet unknown mechanisms. Using biochemical and biophysical approaches, we demonstrate that the trans-membrane domain (TMD) of the HIV-1 envelope (ENV) directly interacts with TLR2 TMD within the membrane milieu. This interaction attenuates TNFα, IL-6 and MCP-1 secretion in macrophages, induced by natural ligands of TLR2 both in in vitro and in vivo models. This was associated with decreased levels of ERK phosphorylation. Furthermore, mutagenesis demonstrated the importance of a conserved GxxxG motif in driving this interaction within the membrane milieu. The administration of the ENV TMD in vivo to lipotechoic acid (LTA)/Galactosamine-mediated septic mice resulted in a significant decrease in mortality and in tissue damage, due to the weakening of systemic macrophage activation. Our findings suggest that the TMD of ENV is involved in modulation of the innate immune response during HIV infection. Furthermore, due to the high functional homology of viral ENV proteins this function may be a general character of viral-induced immune modulation.


Asunto(s)
VIH-1/inmunología , Evasión Inmune/inmunología , Receptor Toll-Like 2/metabolismo , Proteínas del Envoltorio Viral/inmunología , Proteínas del Envoltorio Viral/metabolismo , Animales , Western Blotting , Línea Celular , Dimerización , Femenino , Transferencia Resonante de Energía de Fluorescencia , Infecciones por VIH/inmunología , VIH-1/metabolismo , Inmunidad Innata/inmunología , Activación de Macrófagos/fisiología , Macrófagos/inmunología , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Estructura Terciaria de Proteína/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA