Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
PLoS Pathog ; 18(5): e1010335, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35622876

RESUMEN

Macrophages (MΦ) are increasingly recognized as HIV-1 target cells involved in the pathogenesis and persistence of infection. Paradoxically, in vitro infection assays suggest that virus isolates are mostly T-cell-tropic and rarely MΦ-tropic. The latter are assumed to emerge under CD4+ T-cell paucity in tissues such as the brain or at late stage when the CD4 T-cell count declines. However, assays to qualify HIV-1 tropism use cell-free viral particles and may not fully reflect the conditions of in vivo MΦ infection through cell-to-cell viral transfer. Here, we investigated the capacity of viruses expressing primary envelope glycoproteins (Envs) with CCR5 and/or CXCR4 usage from different stages of infection, including transmitted/founder Envs, to infect MΦ by a cell-free mode and through cell-to-cell transfer from infected CD4+ T cells. The results show that most viruses were unable to enter MΦ as cell-free particles, in agreement with the current view that non-M-tropic viruses inefficiently use CD4 and/or CCR5 or CXCR4 entry receptors on MΦ. In contrast, all viruses could be effectively cell-to-cell transferred to MΦ from infected CD4+ T cells. We further showed that viral transfer proceeded through Env-dependent cell-cell fusion of infected T cells with MΦ targets, leading to the formation of productively infected multinucleated giant cells. Compared to cell-free infection, infected T-cell/MΦ contacts showed enhanced interactions of R5 M- and non-M-tropic Envs with CD4 and CCR5, resulting in a reduced dependence on receptor expression levels on MΦ for viral entry. Altogether, our results show that virus cell-to-cell transfer overcomes the entry block of isolates initially defined as non-macrophage-tropic, indicating that HIV-1 has a more prevalent tropism for MΦ than initially suggested. This sheds light into the role of this route of virus cell-to-cell transfer to MΦ in CD4+ T cell rich tissues for HIV-1 transmission, dissemination and formation of tissue viral reservoirs.


Asunto(s)
Infecciones por VIH , VIH-1 , Antígenos CD4/metabolismo , Linfocitos T CD4-Positivos , Infecciones por VIH/metabolismo , VIH-1/metabolismo , Humanos , Macrófagos/metabolismo , Receptores CCR5/metabolismo , Internalización del Virus
2.
Int J Mol Sci ; 21(24)2020 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-33348900

RESUMEN

Cell-cell fusion between eukaryotic cells is a general process involved in many physiological and pathological conditions, including infections by bacteria, parasites, and viruses. As obligate intracellular pathogens, viruses use intracellular machineries and pathways for efficient replication in their host target cells. Interestingly, certain viruses, and, more especially, enveloped viruses belonging to different viral families and including human pathogens, can mediate cell-cell fusion between infected cells and neighboring non-infected cells. Depending of the cellular environment and tissue organization, this virus-mediated cell-cell fusion leads to the merge of membrane and cytoplasm contents and formation of multinucleated cells, also called syncytia, that can express high amount of viral antigens in tissues and organs of infected hosts. This ability of some viruses to trigger cell-cell fusion between infected cells as virus-donor cells and surrounding non-infected target cells is mainly related to virus-encoded fusion proteins, known as viral fusogens displaying high fusogenic properties, and expressed at the cell surface of the virus-donor cells. Virus-induced cell-cell fusion is then mediated by interactions of these viral fusion proteins with surface molecules or receptors involved in virus entry and expressed on neighboring non-infected cells. Thus, the goal of this review is to give an overview of the different animal virus families, with a more special focus on human pathogens, that can trigger cell-cell fusion.


Asunto(s)
Fusión Celular , Membrana Celular/metabolismo , Fusión de Membrana , Proteínas Virales de Fusión/metabolismo , Internalización del Virus , Virus/metabolismo , Animales , Humanos , Virus/aislamiento & purificación
3.
J Virol ; 91(24)2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-28978713

RESUMEN

HIV-1-infected macrophages participate in virus dissemination and establishment of virus reservoirs in host tissues, but the mechanisms for virus cell-to-cell transfer to macrophages remain unknown. Here, we reveal the mechanisms for cell-to-cell transfer from infected T cells to macrophages and virus spreading between macrophages. We show that contacts between infected T lymphocytes and macrophages lead to cell fusion for the fast and massive transfer of CCR5-tropic viruses to macrophages. Through the merge of viral material between T cells and macrophages, these newly formed lymphocyte-macrophage fused cells acquire the ability to fuse with neighboring noninfected macrophages. Together, these two-step envelope-dependent cell fusion processes lead to the formation of highly virus-productive multinucleated giant cells reminiscent of the infected multinucleated giant macrophages detected in HIV-1-infected patients and simian immunodeficiency virus-infected macaques. These mechanisms represent an original mode of virus transmission for viral spreading and a new model for the formation of macrophage virus reservoirs during infection.IMPORTANCE We reveal a very efficient mechanism involved in cell-to-cell transfer from infected T cells to macrophages and subsequent virus spreading between macrophages by a two-step cell fusion process. Infected T cells first establish contacts and fuse with macrophage targets. The newly formed lymphocyte-macrophage fused cells then acquire the ability to fuse with surrounding uninfected macrophages, leading to the formation of infected multinucleated giant cells that can survive for a long time, as evidenced in vivo in lymphoid organs and the central nervous system. This route of infection may be a major determinant for virus dissemination and the formation of macrophage virus reservoirs in host tissues during HIV-1 infection.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Células Gigantes/virología , Infecciones por VIH/inmunología , VIH-1/fisiología , Macrófagos/citología , Animales , Linfocitos T CD4-Positivos/virología , Fusión Celular , Línea Celular , Células Gigantes/citología , Células HEK293 , VIH-1/patogenicidad , Humanos , Células Jurkat , Macaca mulatta , Macrófagos/virología , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Virus de la Inmunodeficiencia de los Simios/fisiología
4.
J Virol ; 89(6): 3049-61, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25552713

RESUMEN

UNLABELLED: Recently, interferon-induced transmembrane proteins (IFITMs) have been identified to be key effector molecules in the host type I interferon defense system. The invasion of host cells by a large range of RNA viruses is inhibited by IFITMs during the entry step. However, the roles of IFITMs in DNA virus infections have not been studied in detail. In this study, we report that human cytomegalovirus (HCMV), a large human DNA virus, exploits IFITMs to facilitate the formation of the virion assembly compartment (vAC) during infection of human fibroblasts. We found that IFITMs were expressed constitutively in human embryonic lung fibroblasts (MRC5 cells). HCMV infection inhibited IFITM protein accumulation in the later stages of infection. Overexpression of an IFITM protein in MRC5 cells slightly enhanced HCMV production and knockdown of IFITMs by RNA interference reduced the virus titer by about 100-fold on day 8 postinfection, according to the findings of a virus yield assay at a low multiplicity of infection. Virus gene expression and DNA synthesis were not affected, but the typical round structure of the vAC was not formed after the suppression of IFITMs, thereby resulting in defective virion assembly and the production of less infectious virion particles. Interestingly, the replication of herpes simplex virus, a human herpesvirus that is closely related to HCMV, was not affected by the suppression of IFITMs in MRC5 cells. These results indicate that IFITMs are involved in a specific pathway required for HCMV replication. IMPORTANCE: HCMV is known to repurpose the interferon-stimulated genes (ISGs) viperin and tetherin to facilitate its replication. Our results expand the range of ISGs that can be exploited by HCMV for its replication. This is also the first report of a proviral function of IFITMs in DNA virus replication. In addition, whereas previous studies showed that IFITMs modulate virus entry, which is a very early stage in the virus life cycle, we identified a new function of IFITMs during the very late stage of virus replication, i.e., virion assembly. Virus entry and assembly both involve vesicle transport and membrane fusion; thus, a common biochemical activity of IFITMs is likely to be involved. Therefore, our findings may provide a new platform for dissecting the molecular mechanism of action of IFITMs during the blocking or enhancement of virus infection, which are under intense investigation in this field.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Infecciones por Citomegalovirus/metabolismo , Citomegalovirus/crecimiento & desarrollo , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/metabolismo , Virión/crecimiento & desarrollo , Ensamble de Virus , Antígenos de Diferenciación/genética , Línea Celular , Citomegalovirus/genética , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/virología , Interacciones Huésped-Patógeno , Humanos , Proteínas de la Membrana/genética , Proteínas de Unión al ARN/genética , Virión/genética , Virión/fisiología , Replicación Viral
5.
Talanta ; 212: 120749, 2020 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-32113532

RESUMEN

Pb2+ is one of the major environmental pollutants, which can be visually detected by surface plasmon resonance of nanoparticles. Paper based analytical device, as a newly developed microfluidic detection platform, is featured in cost-effective and suitable for on-site analysis. In this paper, a sensitive and portable detection method for Pb2+ was proposed, in which Pb2+ was electrokinetically stacked on the paper fluidic channel by geometric field amplification effect and visualized online by glutathione-modified silver nanoparticles. Colorimetric quantification of the visualized stacking band was conducted by smart phone camera. To avoid unfavorable influence from pH change on the surface plasmon resonance visualization, field amplification effect was introduced by geometric design of the paper fluidic channel. The enriched Pb2+ was clearly visible on the paper substrate, and the stacking band intensity was about four orders of magnitude enhanced, comparing to the intensity without stacking. A linear response to Pb2+ was observed in the range of 0.3-7.0 µM (R2 = 0.997) with a limit of detection of 86 nM and a limit of quantity of 0.28 µM. The established method was used in the detection of Pb2+ from river and lake water samples, and the results were confirmed by atomic absorption spectroscopy method.

6.
Lab Chip ; 19(5): 845-850, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30706062

RESUMEN

On-line enrichment is effective for improving the sensitivity of paper-based analytical devices (PADs). Electrokinetic stacking of ionic species - anionic or cationic species, respectively, on a paper-based fluidic channel has been well demonstrated in the literature. In this work, we further demonstrated that both anionic and cationic species can be electrokinetically stacked and separated simultaneously on the same paper fluidic channel. The feasibility of the proposed method was visually demonstrated by using a colored cationic probe of Rhodamine 6G and an anionic probe of Brilliant Blue. With the introduction of a background electrolyte (BGE) consisting of weak acid and weak base salt, two electric field gradients can be developed on the same paper fluidic channel when a DC voltage was applied. Both of the anionic and cationic species from the reservoirs can be simultaneously stacked as separate bands on the two field gradients, respectively. Under optimized conditions, two orders of magnitude enrichment factors can be achieved for the anionic and cationic probes as characterized by colorimetric analysis by smartphone imaging. The applicability of this method was further demonstrated by stacking and separation of copper ions/nitrite and even amphoteric ions-proteins of phycocyanin (blue, pI 4.4)/cytochrome C (brown, pI 10.2). Potential applications can be found not only for a PAD based point of care test (POCT), but also for sample pretreatment in protein analysis considering the friendliness of the BGE to the mass spectrometer.

7.
mBio ; 10(6)2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31744918

RESUMEN

Dendritic cells (DCs) and macrophages as well as osteoclasts (OCs) are emerging as target cells of HIV-1 involved in virus transmission, dissemination, and establishment of persistent tissue virus reservoirs. While these myeloid cells are poorly infected by cell-free viruses because of the high expression levels of cellular restriction factors such as SAMHD1, we show here that HIV-1 uses a specific and common cell-to-cell fusion mechanism for virus transfer and dissemination from infected T lymphocytes to the target cells of the myeloid lineage, including immature DCs (iDCs), OCs, and macrophages, but not monocytes and mature DCs. The establishment of contacts with infected T cells leads to heterotypic cell fusion for the fast and massive transfer of viral material into OC and iDC targets, which subsequently triggers homotypic fusion with noninfected neighboring OCs and iDCs for virus dissemination. These two cell-to-cell fusion processes are not restricted by SAMHD1 and allow very efficient spreading of virus in myeloid cells, resulting in the formation of highly virus-productive multinucleated giant cells. These results reveal the cellular mechanism for SAMHD1-independent cell-to-cell spreading of HIV-1 in myeloid cell targets through the formation of the infected multinucleated giant cells observed in vivo in lymphoid and nonlymphoid tissues of HIV-1-infected patients.IMPORTANCE We demonstrate that HIV-1 uses a common two-step cell-to-cell fusion mechanism for massive virus transfer from infected T lymphocytes and dissemination to myeloid target cells, including dendritic cells and macrophages as well as osteoclasts. This cell-to-cell infection process bypasses the restriction imposed by the SAMHD1 host cell restriction factor for HIV-1 replication, leading to the formation of highly virus-productive multinucleated giant cells as observed in vivo in lymphoid and nonlymphoid tissues of HIV-1-infected patients. Since myeloid cells are emerging as important target cells of HIV-1, these results contribute to a better understanding of the role of these myeloid cells in pathogenesis, including cell-associated virus sexual transmission, cell-to-cell virus spreading, and establishment of long-lived viral tissue reservoirs.


Asunto(s)
Infecciones por VIH/metabolismo , Infecciones por VIH/virología , VIH-1/fisiología , Proteína 1 que Contiene Dominios SAM y HD/metabolismo , Tropismo Viral , Replicación Viral , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/virología , Células Dendríticas/metabolismo , Células Dendríticas/virología , Humanos , Macrófagos/metabolismo , Macrófagos/virología , Células Mieloides/metabolismo , Células Mieloides/virología
8.
Front Immunol ; 9: 260, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29515578

RESUMEN

While HIV-1 infection of target cells with cell-free viral particles has been largely documented, intercellular transmission through direct cell-to-cell contact may be a predominant mode of propagation in host. To spread, HIV-1 infects cells of the immune system and takes advantage of their specific particularities and functions. Subversion of intercellular communication allows to improve HIV-1 replication through a multiplicity of intercellular structures and membrane protrusions, like tunneling nanotubes, filopodia, or lamellipodia-like structures involved in the formation of the virological synapse. Other features of immune cells, like the immunological synapse or the phagocytosis of infected cells are hijacked by HIV-1 and used as gateways to infect target cells. Finally, HIV-1 reuses its fusogenic capacity to provoke fusion between infected donor cells and target cells, and to form infected syncytia with high capacity of viral production and improved capacities of motility or survival. All these modes of cell-to-cell transfer are now considered as viral mechanisms to escape immune system and antiretroviral therapies, and could be involved in the establishment of persistent virus reservoirs in different host tissues.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Sinapsis Inmunológicas/virología , Uniones Intercelulares/virología , Animales , Linfocitos T CD4-Positivos/virología , Reservorios de Enfermedades , Infecciones por VIH/transmisión , Humanos , Evasión Inmune , Fusión de Membrana , Nanotubos/virología , Seudópodos/virología
9.
Sci Rep ; 6: 26167, 2016 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-27188239

RESUMEN

Functional analysis of the essential proteins encoded by human cytomegalovirus (HCMV) is hindered by the lack of complementing systems. To overcome this difficulty, we have established a novel approach, termed the intein-mediated modulation of protein stability (imPS), in which a destabilizing domain and part of a split intein are fused to the essential protein. The growth of the mutant virus can then be regulated by the degradation and splicing of the protein. We found that an ultrafast gp41-1 split intein was able to rescue or degrade the protein of interest (POI) by removing or adding a strong degron through protein splicing. As a result, the function of the POI was turned on or off during the process. Using HCMV essential gene IE1/IE2, we confirmed that imPS worked remarkably well in conditionally regulating protein stability during viral infection. This conditional approach is likely to be applicable for dissecting the gene functions of HCMV or other viruses.


Asunto(s)
Citomegalovirus/genética , Genes Esenciales , Genes Virales , Biología Molecular/métodos , Virología/métodos , Citomegalovirus/crecimiento & desarrollo , Células HEK293 , Humanos , Inteínas/genética , Viabilidad Microbiana , Estabilidad Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA