Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurochem ; 163(4): 338-356, 2022 11.
Article in English | MEDLINE | ID: mdl-36205031

ABSTRACT

Human immunodeficiency virus (HIV) neuroinvasion occurs early after infection through the trafficking of virus-infected immune cells into the central nervous system (CNS) and viral dissemination into the brain. There, it can infect resident brain cells including astrocytes, the most abundant cell type that is crucial to brain homeostasis. In this report, we examined the HIV-related mechanism able to induce bystander cell death in astrocytes mediated by cell-to-cell contact with productively infected (PI) ones. We first demonstrate that HIV-induced bystander cell death involves mitochondrial dysfunction that promotes exacerbated reactive oxygen species production. Such a phenomenon is a contagious cell death that requires contact with HIV-PI astrocytes that trigger caspase-dependent (apoptosis and pyroptosis) and caspase-independent cell death pathways. The HIV accessory proteins Nef, Vpu, and Vpr counteract astrocyte death among PI cells but, in contrast, participate to promote contagious bystander cell death by inducing mitochondrial reactive oxygen species production. Our findings indicate that astrocytes PI by HIV became capable to counteract infection-derived death signals, surviving, and spreading the bystander cell death into neighboring uninfected cells by a cell-to-cell contact-dependent mechanism. Considering that astrocytes have been proposed as a long-term HIV reservoir in the CNS, ascertaining the mechanism of survival and contagious bystander death will afford clear targets in the current goal to achieve a functional cure.


Subject(s)
HIV Infections , HIV-1 , Humans , Astrocytes/metabolism , HIV-1/physiology , Reactive Oxygen Species/metabolism , Cell Death , Caspases/metabolism
2.
Front Microbiol ; 11: 563320, 2020.
Article in English | MEDLINE | ID: mdl-33193149

ABSTRACT

Introduction: Trypanosoma cruzi is an intracellular protozoa and etiological agent that causes Chagas disease. Its presence among the immunocompromised HIV-infected individuals is relevant worldwide because of its impact on the central nervous system (CNS) causing severe meningoencephalitis. The HIV infection of astrocytes - the most abundant cells in the brain, where the parasite can also be hosted - being able to modify reactive oxygen species (ROS) could influence the parasite growth. In such interaction, extracellular vesicles (EVs) shed from trypomastigotes may alter the surrounding environment including its pro-oxidant status. Methods: We evaluated the interplay between both pathogens in human astrocytes and its consequences on the host cell pro-oxidant condition self-propitiated by the parasite - using its EVs - or by HIV infection. For this goal, we challenged cultured human primary astrocytes with both pathogens and the efficiency of infection and multiplication were measured by microscopy and flow cytometry and parasite DNA quantification. Mitochondrial and cellular ROS levels were measured by flow cytometry in the presence or not of scavengers with a concomitant evaluation of the cellular apoptosis level. Results: We observed that increased mitochondrial and cellular ROS production boosted significantly T. cruzi infection and multiplication in astrocytes. Such oxidative condition was promoted by free trypomastigotes-derived EVs as well as by HIV infection. Conclusions: The pathogenesis of the HIV-T. cruzi coinfection in astrocytes leads to an oxidative misbalance as a key mechanism, which exacerbates ROS generation and promotes positive feedback to parasite growth in the CNS.

3.
Arch Virol ; 165(12): 2915-2919, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32978684

ABSTRACT

Human immunodeficiency virus type 1 (HIV) primary drug resistance mutations (DRMs) influence the long-term therapeutic effects of antiretroviral treatment (ART). Drug-resistance genotyping based on polymerase gene sequences obtained by next-generation sequencing (NGS) was performed using samples from 10 ART-naïve HIV-infected men who have sex with men (MSM; P1-P10) from the acute/early to chronic stage of infection. Three of the 10 subjects exhibited the presence of major (abundance, ≥ 20%) viral populations carrying DRM at early/acute stage that later, at the chronic stage, dropped drastically (V106M) or remained highly abundant (E138A). Four individuals exhibited additional DRMs (M46I/L; I47A; I54M, L100V) as HIV minority populations (abundance, 2-20%) that emerged during the chronic stage but ephemerally.


Subject(s)
Anti-HIV Agents/pharmacology , Drug Resistance, Viral/genetics , HIV Infections/virology , HIV-1/genetics , Mutation , HIV Infections/drug therapy , HIV-1/drug effects , High-Throughput Nucleotide Sequencing , Homosexuality, Male , Humans , Male , Phylogeny , Sexual and Gender Minorities , Viral Load
4.
Front Immunol ; 9: 2633, 2018.
Article in English | MEDLINE | ID: mdl-30515154

ABSTRACT

Despite more than 30 years of extensive research efforts, a complete understanding of the neurological consequences of HIV central nervous system (CNS) infection remains elusive. HIV is not only able to establish a viral reservoir in the CNS but also to initiate manifestation of neurodegenerative diseases. These neurological disorders may arise because of virus-induced activation of the inflammasome in CNS cells, including astrocytes. Nevertheless, in some productive viral infection scenarios, selective autophagy may reduce inflammation through mitochondrial degradation ("mitophagy") to counteract inflammasome activation. In this study, using cultured human astrocytes, we demonstrate that-depending on the HIV infection outcome-cells may resist death, or succumb by inflammasome activation when viral infection is productive or abortive, respectively. Cells productively infected with HIV were able to attenuate both mitochondrial ROS production and mitochondrial membrane potential dissipation, thus exhibiting cell death resistance. Interestingly, mitochondrial injury was counteracted by increasing the autophagic flux and by activating mitophagy. Conversely, astrocytes exposed to HIV in an abortive scenario showed prominent mitochondrial damage, inflammasome activation, and cell death. This bystander effect occurred after cell-to-cell contact with HIV-productively infected astrocytes. In summary, we demonstrate a tight functional crosstalk between viral infection mode, inflammasome activation, autophagy pathways and cell fate in the context of HIV infection. Moreover, mitophagy is crucial for cell death resistance in HIV-productively infected astrocytes, but its impairment may favor inflammasome-mediated cell death in abortively infected cells.


Subject(s)
Astrocytes/immunology , Bystander Effect/immunology , HIV Infections/immunology , HIV-1/immunology , Inflammasomes/immunology , Mitophagy/immunology , Astrocytes/pathology , Cell Death/immunology , HIV Infections/pathology , Humans
5.
Article in English | MEDLINE | ID: mdl-28824880

ABSTRACT

The protozoan Trypanosoma cruzi is the etiological agent of Chagas disease. In immunosuppressed individuals, as it occurs in the coinfection with human immunodeficiency virus (HIV), the central nervous system may be affected. In this regard, reactivation of Chagas disease is severe and often lethal, and it accounts for meningoencephalitis. Astrocytes play a crucial role in the environment maintenance of healthy neurons; however, they can host HIV and T. cruzi. In this report, human astrocytes were infected in vitro with both genetically modified-pathogens to express alternative fluorophore. As evidenced by fluorescence microscopy and flow cytometry, HIV and T. cruzi coexist in the same astrocyte, likely favoring reciprocal interactions. In this context, lower rates of cell death were observed in both T. cruzi monoinfected-astrocytes and HIV-T. cruzi coinfection in comparison with those infected only with HIV. The level of HIV replication is significantly diminished under T. cruzi coinfection, but without affecting the infectivity of the HIV progeny. This interference with viral replication appears to be related to the T. cruzi multiplication rate or its increased intracellular presence but does not require their intracellular cohabitation or infected cell-to-cell contact. Among several Th1/Th2/Th17 profile-related cytokines, only IL-6 was overexpressed in HIV-T. cruzi coinfection exhibiting its cytoprotective role. This study demonstrates that T. cruzi and HIV are able to coinfect astrocytes thus altering viral replication and apoptosis.


Subject(s)
Apoptosis , Astrocytes/immunology , Chagas Disease/complications , Coinfection , HIV Infections/complications , Virus Replication/physiology , Apoptosis/drug effects , Astrocytes/parasitology , Astrocytes/virology , Cell Death , Cell Line , Chagas Disease/immunology , Chagas Disease/virology , Cytokines/metabolism , HIV/physiology , HIV Infections/immunology , Herpesvirus 2, Human/physiology , Humans , Interleukin-6 , Nitroimidazoles/pharmacology , Th1 Cells/immunology , Th17 Cells/immunology , Th2 Cells/immunology , Trypanosoma cruzi/genetics , Trypanosoma cruzi/immunology , Trypanosoma cruzi/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...