Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Neuroinflammation ; 20(1): 184, 2023 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-37537664

RESUMEN

BACKGROUND: Although mainly causing a respiratory syndrome, numerous neurological symptoms have been identified following of SARS-CoV-2 infection. However, how the virus affects the brain and how the mutations carried by the different variants modulate those neurological symptoms remain unclear. METHODS: We used primary human pericytes, foetal astrocytes, endothelial cells and a microglial cell line to investigate the effect of several SARS-CoV-2 variants of concern or interest on their functional activities. Cells and a 3D blood-brain barrier model were infected with the wild-type form of SARS-CoV-2, Alpha, Beta, Delta, Eta, or Omicron (BA.1) variants at various MOI. Cells and supernatant were used to evaluate cell susceptibility to the virus using a microscopic assay as well as effects of infection on (i) cell metabolic activity using a colorimetric MTS assay; (ii) viral cytopathogenicity using the xCELLigence system; (iii) extracellular glutamate concentration by fluorometric assay; and (iv) modulation of blood-brain barrier permeability. RESULTS: We demonstrate that productive infection of brain cells is SARS-CoV-2 variant dependent and that all the variants induce stress to CNS cells. The wild-type virus was cytopathic to all cell types except astrocytes, whilst Alpha and Beta variants were only cytopathic for pericytes, and the Omicron variant cytopathic for endothelial cells and pericytes. Lastly wild-type virus increases blood-brain barrier permeability and all variants, except Beta, modulate extracellular glutamate concentration, which can lead to excitotoxicity or altered neurotransmission. CONCLUSIONS: These results suggest that SARS-CoV-2 is neurotropic, with deleterious consequences for the blood-brain barrier integrity and central nervous system cells, which could underlie neurological disorders following SARS-CoV-2 infection.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Barrera Hematoencefálica , Células Endoteliales , Ácido Glutámico
2.
Glia ; 68(11): 2212-2227, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32250524

RESUMEN

Since the introduction of the combined antiretroviral therapy, HIV-1 infection has become a manageable chronic disease in which patients display a life expectancy almost identical to the general population. Nevertheless, various age-related pathologies such as neurocognitive disorders have emerged as serious complications. A "shock and kill" strategy using latency-reversing agents (LRA) to reactivate HIV-1 has been proposed to eliminate the viral reservoir in such chronically infected patients. However, the impact of LRA on the central nervous system remains elusive. Given that an increased amyloid beta (Aß) deposition is a feature of HIV-1-infected brains, we investigated the consequences of HIV-1 infection and treatment with two LRA (bryostatin-1 and JQ1) on the capacity of human astrocytes to engulf and clear Aß. We show here that HIV-1-infected astrocytes accumulate a very high amount of Aß compared to uninfected cells, but the engulfed peptide in degraded very slowly. The LRA bryostatin-1 induces a reduction in Aß endocytosis, whereas JQ1 treatment results in a very slow degradation of the ingested material associated with a reduced expression of the endopeptidase neprilysin. An exposure to JQ1 also induces a sustained release of Aß-loaded microvesicles. Thus, both HIV-1 infection and treatment with some LRA could contribute to the reported Aß accumulation in the brain of HIV-1-infected persons.


Asunto(s)
Infecciones por VIH , VIH-1 , Péptidos beta-Amiloides , Astrocitos , Azepinas , Brioestatinas/farmacología , Infecciones por VIH/tratamiento farmacológico , Homeostasis , Humanos , Triazoles , Activación Viral , Latencia del Virus
3.
J Immunol ; 198(3): 1229-1241, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27994072

RESUMEN

A shock-and-kill approach involving the simultaneous treatment of HIV-1-infected patients with latency-reversing agents (LRAs) and combination antiretroviral therapy was proposed as a means to eradicate viral reservoirs. Currently available LRAs cannot discriminate between HIV-1-infected and uninfected cells. Therefore, the risks and benefits of using broad-spectrum LRAs need to be carefully evaluated, particularly in the CNS, where inflammation and leukocyte transmigration must be tightly regulated. We used a real-time impedance-sensing system to dynamically record the impact of different classes of LRAs on the integrity of tight monolayers of the immortalized human cerebral microvascular endothelial cell line hCMEC/D3. Results show that prostratin and bryostatin-1 can significantly damage the integrity of an endothelial monolayer. Moreover, prostratin and bryostatin-1 induce secretion of some proinflammatory cytokines and an increase of ICAM-1 expression. Additional studies demonstrated that prostratin and bryostatin-1 also affect adhesion and transmigration of CD4+ and CD8+ T cells as well as monocytes in an in vitro human blood-brain barrier (BBB) model. Prostratin and bryostatin-1 could thus be considered as potent regulators of BBB permeability and inflammation that influence leukocyte transport across the BBB. Altogether, these findings contribute to a better understanding of the potential risks and benefits of using a shock-and-kill approach with LRAs on the normal physiological functions of the BBB.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Brioestatinas/farmacología , VIH-1/fisiología , Leucocitos/efectos de los fármacos , Ésteres del Forbol/farmacología , Latencia del Virus/efectos de los fármacos , Acetamidas/farmacología , Azacitidina/análogos & derivados , Azacitidina/farmacología , Azepinas/farmacología , Brioestatinas/efectos adversos , Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/análisis , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CCL2/fisiología , Citocinas/metabolismo , Decitabina , Humanos , Inflamación/inducido químicamente , Molécula 1 de Adhesión Intercelular/análisis , Leucocitos/fisiología , Ésteres del Forbol/efectos adversos , Quinazolinas/farmacología , Receptores de Superficie Celular/análisis
4.
Glia ; 66(7): 1363-1381, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29464785

RESUMEN

The "shock and kill" HIV-1 cure strategy proposes eradication of stable cellular reservoirs by clinical treatment with latency-reversing agents (LRAs). Although resting CD4+ T cells latently infected with HIV-1 constitute the main reservoir that is targeted by these approaches, their consequences on other reservoirs such as the central nervous system are still unknown and should be taken into consideration. We performed experiments aimed at defining the possible role of astrocytes in HIV-1 persistence in the brain and the effect of LRA treatments on this viral sanctuary. We first demonstrate that the diminished HIV-1 production in a proliferating astrocyte culture is due to a reduced proliferative capacity of virus-infected cells compared with uninfected astrocytes. In contrast, infection of non-proliferating astrocytes led to a robust HIV-1 infection that was sustained for over 60 days. To identify astrocytes latently infected with HIV-1, we designed a new dual-color reporter virus called NL4.3 eGFP-IRES-Crimson that is fully infectious and encodes for all viral proteins. Although we detected a small fraction of astrocytes carrying silent HIV-1 proviruses, we did not observe any reactivation using various LRAs and even strong inducers such as tumor necrosis factor, thus suggesting that these proviruses were either not transcriptionally competent or in a state of deep latency. Our findings imply that astrocytes might not constitute a latent reservoir per se but that relentless virus production by this brain cell population could contribute to the neurological disorders seen in HIV-1-infected persons subjected to combination antiretroviral therapy.


Asunto(s)
Astrocitos/fisiología , Astrocitos/virología , Infecciones por VIH/fisiopatología , VIH-1/fisiología , Astrocitos/patología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD4-Positivos/virología , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Células HEK293 , VIH-1/genética , Humanos , Latencia del Virus
5.
J Neuroinflammation ; 14(1): 242, 2017 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-29228979

RESUMEN

BACKGROUND: Despite effectiveness of the combined antiretroviral therapy, HIV-1 persists in long-lived latently infected cells. Consequently, new therapeutic approaches aimed at eliminating this latent reservoir are currently being developed. A "shock and kill" strategy using latency-reversing agents (LRA) to reactivate HIV-1 has been proposed. However, the impact of LRA on the central nervous system (CNS) remains elusive. METHODS: We used human fetal astrocytes and investigated the effects of several LRA on their functional and secretory activities. Astrocytes were infected with VSV-G-pseudotyped HIV-1 before treatment with various blood-brain barrier (BBB)-permeable LRA at subcytotoxic doses, which allow HIV-1 reactivation based on previous in vitro and clinical studies. Cells and supernatants were then used to evaluate effects of infection and LRA on (i) viability and metabolic activity of astrocytes using a colorimetric MTS assay; (ii) chemokines and proinflammatory cytokines secretion and gene expression by astrocytes using ELISA and RT-qPCR, respectively; (iii) expression of complement component 3 (C3), a proxy for astrogliosis, by RT-qPCR; (iv) glutamate uptake capacity by a fluorometric assay; and (v) modulation of neutrophil transmigration across an in vitro BBB model. RESULTS: We demonstrate that bryostatin-1 induces secretion of chemokines CCL2 and IL-8 and proinflammatory cytokines IL-6 and GM-CSF, whereas their production is repressed by JQ1. Bryostatin-1 also increases expression of complement component 3 and perturbs astrocyte glutamate homeostasis. Lastly, bryostatin-1 enhances transmigration of neutrophils across an in vitro blood-brain barrier model and induces formation of neutrophil extracellular traps. CONCLUSIONS: These observations highlight the need to carefully assess the potential harmful effect to the CNS when selecting LRA for HIV-1 reactivation strategies.


Asunto(s)
Adyuvantes Inmunológicos/toxicidad , Astrocitos/efectos de los fármacos , Azepinas/toxicidad , Encéfalo/efectos de los fármacos , Brioestatinas/toxicidad , Quimiotaxis de Leucocito/efectos de los fármacos , Triazoles/toxicidad , Encéfalo/patología , VIH-1/fisiología , Humanos , Inflamación/patología , Neutrófilos/efectos de los fármacos , Activación Viral/efectos de los fármacos , Latencia del Virus/efectos de los fármacos
6.
J Virol ; 88(9): 5109-21, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24574390

RESUMEN

UNLABELLED: Human immunodeficiency virus type 1 (HIV-1) replication in dendritic cells (DCs) is restricted by SAMHD1. This factor is counteracted by the viral protein Vpx; Vpx is found in HIV-2 and simian immunodeficiency virus (SIV) from sooty mangabeys (SIVsm) or from macaques (SIVmac) but is absent from HIV-1. We previously observed that HIV-1 replication in immature DCs is stimulated by cocultivation with primary T and B lymphocytes, suggesting that HIV-1 restriction in DCs may be overcome under coculture conditions. Here, we aimed to decipher the mechanism of SAMHD1-mediated restriction in DC-lymphocyte coculture. We found that coculture with lymphocytes downregulated SAMHD1 expression and was associated with increased HIV-1 replication in DCs. Moreover, in infected DC-T lymphocyte cocultures, DCs acquired maturation status and secreted type 1 interferon (alpha interferon [IFN-α]). The blockade of DC-lymphocyte cross talk by anti-ICAM-1 antibody markedly inhibited the stimulation of HIV-1 replication and prevented the downregulation of SAMHD1 expression in cocultured DCs. These results demonstrate that, in contrast to purified DCs, cross talk with lymphocytes downregulates SAMHD1 expression in DCs, triggering HIV-1 replication and an antiviral immune response. Therefore, HIV-1 replication and immune sensing by DCs should be investigated in more physiologically relevant models of DC/lymphocyte coculture. IMPORTANCE: SAMHD1 restricts HIV-1 replication in dendritic cells (DCs). Here, we demonstrate that, in a coculture model of DCs and lymphocytes mimicking early mucosal HIV-1 infection, stimulation of HIV-1 replication in DCs is associated with downregulation of SAMHD1 expression and activation of innate immune sensing by DCs. We propose that DC-lymphocyte cross talk occurring in vivo modulates host restriction factor SAMHD1, promoting HIV-1 replication in cellular reservoirs and stimulating immune sensing.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/virología , VIH-1/fisiología , Linfocitos/inmunología , Proteínas de Unión al GTP Monoméricas/biosíntesis , Replicación Viral , Técnicas de Cocultivo , Regulación hacia Abajo , Humanos , Proteína 1 que Contiene Dominios SAM y HD , Cultivo de Virus
7.
Blood ; 120(18): 3708-17, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22955927

RESUMEN

Dendritic cells (DCs) support only low levels of HIV-1 replication, but have been shown to transfer infectious viral particles highly efficiently to neighboring permissive CD4 T lymphocytes. This mode of cell-to-cell HIV-1 spread may be a predominant mode of infection and dissemination. In the present study, we analyzed the kinetics of fusion, replication, and the ability of HIV-1-specific Abs to inhibit HIV-1 transfer from immature DCs to autologous CD4 T lymphocytes. We found that neutralizing mAbs prevented HIV-1 transfer to CD4 T lymphocytes in trans and in cis, whereas nonneutralizing Abs did not. Neutralizing Abs also significantly decreased HIV-1 replication in DCs, even when added 2 hours after HIV-1 infection. Interestingly, a similar inhibition of HIV-1 replication in DCs was detected with some nonneutralizing Abs and was correlated with DC maturation. We suggest that the binding of HIV-1-specific Abs to FcγRs leads to HIV-1 inhibition in DCs by triggering DC maturation. This efficient inhibition of HIV-1 transfer by Abs highlights the importance of inducing HIV-specific Abs by vaccination directly at the mucosal portal of HIV-1 entry to prevent early dissemination after sexual transmission.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Linfocitos T CD4-Positivos/virología , Células Dendríticas/virología , Infecciones por VIH/transmisión , Infecciones por VIH/virología , Linfocitos T CD4-Positivos/inmunología , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/inmunología , Citometría de Flujo , VIH-1/inmunología , Humanos
8.
Artículo en Inglés | MEDLINE | ID: mdl-38988046

RESUMEN

Conditions affecting the brain are the second leading cause of death globally. One of the main challenges for drugs targeting brain diseases is passing the blood-brain barrier (BBB). Here, the effectiveness of mesoporous silica nanostars (MSiNSs) with two different spike lengths to cross an in vitro BBB multicellular model was evaluated and compared to spherical nanoparticles (MSiNP). A modified sol-gel single-micelle epitaxial growth was used to produce MSiNS, which showed no cytotoxicity or immunogenicity at concentrations of up to 1 µg mL-1 in peripheral blood mononuclear and neuronal cells. The nanostar MSiNS effectively penetrated the BBB model after 24 h, and MSiNS-1 with a shorter spike length (9 ± 2 nm) crossed the in vitro BBB model more rapidly than the MSiNS-2 with longer spikes (18 ± 4 nm) or spherical MSiNP at 96 h, which accumulated in the apical and basolateral sides, respectively. Molecular dynamic simulations illustrated an increase in configurational flexibility of the lipid bilayer during contact with the MSiNS, resulting in wrapping, whereas the MSiNP suppressed membrane fluctuations. This work advances an effective brain drug delivery system based on virus-like shaped MSiNS for the treatment of different brain diseases and a mechanism for their interaction with lipid bilayers.

9.
J Leukoc Biol ; 105(2): 267-280, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30645042

RESUMEN

Tuberculosis (TB) remains a leading cause of death globally. Dissemination of TB to the brain results in the most severe form of extrapulmonary TB, tuberculous meningitis (TBM), which represents a medical emergency associated with high rates of mortality and disability. Via various mechanisms the Mycobacterium tuberculosis (M.tb) bacillus disseminates from the primary site of infection and overcomes protective barriers to enter the CNS. There it induces an inflammatory response involving both the peripheral and resident immune cells, which initiates a cascade of pathologic mechanisms that may either contain the disease or result in significant brain injury. Here we review the steps from primary infection to cerebral disease, factors that contribute to the virulence of the organism and the vulnerability of the host and discuss the immune response and the clinical manifestations arising. Priorities for future research directions are suggested.


Asunto(s)
Tuberculosis Meníngea/etiología , Sistema Nervioso Central/microbiología , Sistema Nervioso Central/patología , Sistema Nervioso Central/fisiopatología , Infecciones por VIH/inmunología , Infecciones por VIH/microbiología , Humanos , Inmunidad , Mycobacterium tuberculosis/patogenicidad , Tuberculosis Meníngea/microbiología , Tuberculosis Meníngea/patología , Tuberculosis Meníngea/fisiopatología , Virulencia
10.
Methods Appl Fluoresc ; 6(3): 037002, 2018 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-29765013

RESUMEN

Extracellular traps ejected by various immune cells (neutrophils, macrophages, eosinophils and mast cells) have several immune functions, either protective against pathogens or deleterious in some autoimmune or inflammatory disorders. Since their first description in 2004, the mechanisms of extracellular traps formation have been extensively investigated though still not fully understood. We describe here a new tool for the detection of extracellular traps by fluorescence microscopy in a single-step staining protocol, which does not require any wash. The approach uses the GreenGlo™ DNA dye, which can differentiate between nuclear DNA and extracellular DNA (extracellular traps) released from cells using different fluorescence excitation wavelengths. GreenGlo™ staining is suitable for adherent and non-adherent cells and is expected to be extendable to extracellular traps from other cells types (i.e. eosinophils, mast cells and monocytes).


Asunto(s)
ADN/genética , Trampas Extracelulares/inmunología , Microscopía Fluorescente/métodos , Humanos
11.
AIDS ; 28(5): 667-77, 2014 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-24451159

RESUMEN

OBJECTIVE: The chronology of HIV infection in mucosal tissue after sexual transmission is unknown. Several potential HIV target cells are present at these sites, including dendritic cells, macrophages, and CD4(+) T lymphocytes. Dendritic cells and macrophages are antigen-presenting cells (APCs) and are thus involved in cross-talk with T cells. This close contact may favor efficient HIV-1 transfer to T lymphocytes, resulting in rapid HIV-1 dissemination. DESIGN: We investigated the role of APCs in HIV transfer to T cells by incubating Langerhans cells and interstitial dendritic cells (IDCs) or monocyte-derived macrophages (MDMs) with HIV for 2 h before addition of uninfected autologous CD4(+) T lymphocytes. METHODS: HIV infection was recorded after different time points. Following staining, the measurement of intracellular p24 in the different cell populations was analyzed by flow cytometry. RESULTS: We showed that Langerhans cells/IDCs and macrophages efficiently transferred HIV to CD4(+) T cells. Interestingly, a rapid HIV transfer in trans predominated in MDMs, whereas cis transfer mainly occurred in Langerhans cells/IDC cocultures. Neutralizing antibody 2G12, added to HIV-loaded APCs, efficiently blocked both the trans and the cis infection of T cells. CONCLUSION: These findings highlight the major contributions of various mucosal cells in HIV dissemination and suggest that HIV hijacks the different properties of APCs to favor its dissemination through the body. They emphasize the role of macrophages in the rapid transmission of HIV to T lymphocytes at mucosal sites, dendritic cells being prone to migration to lymphoid organ for subsequent dissemination by cis transfer.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Células Dendríticas/virología , Infecciones por VIH/virología , VIH-1/aislamiento & purificación , Macrófagos/virología , Células Cultivadas , Técnicas de Cocultivo , Citometría de Flujo , Proteína p24 del Núcleo del VIH/análisis , Humanos , Recién Nacido , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA