Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 566
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(35): e2406421121, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39159381

RESUMEN

Viral infection is frequently assayed by ongoing expression of viral genes. These assays fail to identify cells that have been exposed to the virus but limit or inhibit viral replication. To address this limitation, we used a dual-labeling vesicular stomatitis virus (DL-VSV), which has a deletion of the viral glycoprotein gene, to allow evaluation of primary infection outcomes. This virus encodes Cre, which can stably mark any cell with even a minimal level of viral gene expression. Additionally, the virus encodes GFP, which distinguishes cells with higher levels of viral gene expression, typically due to genome replication. Stereotactic injections of DL-VSV into the murine brain showed that different cell types had very different responses to the virus. Almost all neurons hosted high levels of viral gene expression, while glial cells varied in their responses. Astrocytes (Sox9+) were predominantly productively infected, while oligodendrocytes (Sox10+) were largely abortively infected. Microglial cells (Iba1+) were primarily uninfected. Furthermore, we monitored the early innate immune response to viral infection and identified unique patterns of interferon (IFN) induction. Shortly after infection, microglia were the main producers of IFNb, whereas later, oligodendrocytes were the main producers. IFNb+ cells were primarily abortively infected regardless of cell type. Last, we investigated whether IFN signaling had any impact on the outcome of primary infection and did not observe significant changes, suggesting that intrinsic factors are likely responsible for determining the outcome of primary infection.


Asunto(s)
Astrocitos , Animales , Ratones , Astrocitos/virología , Astrocitos/metabolismo , Replicación Viral , Microglía/virología , Microglía/metabolismo , Microglía/inmunología , Neuronas/virología , Neuronas/metabolismo , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética , Vesiculovirus/fisiología , Vesiculovirus/inmunología , Vesiculovirus/genética , Oligodendroglía/virología , Oligodendroglía/metabolismo , Estomatitis Vesicular/virología , Estomatitis Vesicular/inmunología , Inmunidad Innata , Ratones Endogámicos C57BL , Encéfalo/virología , Encéfalo/metabolismo , Encéfalo/inmunología , Neuroglía/virología , Neuroglía/metabolismo
2.
PLoS Pathog ; 20(9): e1012168, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39283947

RESUMEN

Human Immunodeficiency Virus (HIV) is widely acknowledged for its profound impact on the immune system. Although HIV primarily affects peripheral CD4 T cells, its influence on the central nervous system (CNS) cannot be overlooked. Within the brain, microglia and CNS-associated macrophages (CAMs) serve as the primary targets for HIV and the simian immunodeficiency virus (SIV) in nonhuman primates. This infection can lead to neurological effects and establish a viral reservoir. Given the gaps in our understanding of how these cells respond in vivo to acute CNS infection, we conducted single-cell RNA sequencing (scRNA-seq) on myeloid cells from the brains of three rhesus macaques 12 days after SIV infection, along with three uninfected controls. Our analysis revealed six distinct microglial clusters including homeostatic microglia, preactivated microglia, and activated microglia expressing high levels of inflammatory and disease-related molecules. In response to acute SIV infection, the homeostatic and preactivated microglia population decreased, while the activated and disease-related microglia increased. All microglial clusters exhibited upregulation of MHC class I molecules and interferon-related genes, indicating their crucial roles in defending against SIV during the acute phase. All microglia clusters also upregulated genes linked to cellular senescence. Additionally, we identified two distinct CAM populations: CD14lowCD16hi and CD14hiCD16low CAMs. Interestingly, during acute SIV infection, the dominant CAM population changed to one with an inflammatory phenotype. Specific upregulated genes within one microglia and one macrophage cluster were associated with neurodegenerative pathways, suggesting potential links to neurocognitive disorders. This research sheds light on the intricate interactions between viral infection, innate immune responses, and the CNS, providing valuable insights for future investigations.


Asunto(s)
Macaca mulatta , Macrófagos , Microglía , Síndrome de Inmunodeficiencia Adquirida del Simio , Virus de la Inmunodeficiencia de los Simios , Análisis de la Célula Individual , Animales , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Microglía/inmunología , Microglía/virología , Virus de la Inmunodeficiencia de los Simios/inmunología , Macrófagos/inmunología , Macrófagos/virología , Sistema Nervioso Central/virología , Sistema Nervioso Central/inmunología , Encéfalo/virología , Encéfalo/inmunología , Encéfalo/patología
3.
Rev Med Virol ; 34(4): e2565, 2024 07.
Artículo en Inglés | MEDLINE | ID: mdl-39031738

RESUMEN

Global attention is riveted on neurodegenerative diseases due to their unresolved aetiologies and lack of efficacious therapies. Two key factors implicated include mitochondrial impairment and microglial ageing. Several viral infections, including Herpes simplex virus-1 (HSV-1), human immunodeficiency virus (HIV) and Epstein-Barr virus, are linked to heightened risk of these disorders. Surprisingly, numerous studies indicate viruses induce these aforementioned precipitating events. Epstein-Barr virus, Hepatitis C Virus, HIV, respiratory syncytial virus, HSV-1, Japanese Encephalitis Virus, Zika virus and Enterovirus 71 specifically impact mitochondrial function, leading to mitochondrial malfunction. These vital organelles govern various cell activities and, under specific circumstances, trigger microglial ageing. This article explores the role of viral infections in elucidating the pathogenesis of neurodegenerative ailments. Various viruses instigate microglial ageing via mitochondrial destruction, causing senescent microglia to exhibit activated behaviour, thereby inducing neuroinflammation and contributing to neurodegeneration.


Asunto(s)
Microglía , Mitocondrias , Enfermedades Neurodegenerativas , Virosis , Humanos , Enfermedades Neurodegenerativas/virología , Enfermedades Neurodegenerativas/patología , Mitocondrias/metabolismo , Virosis/virología , Virosis/patología , Microglía/virología , Microglía/patología , Animales
4.
Brain ; 147(9): 3059-3069, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39049445

RESUMEN

Perivascular macrophages (PVMs) and, to a lesser degree, microglia are targets and reservoirs of HIV and simian immunodeficiency virus (SIV) in the brain. Previously, we demonstrated that colony-stimulating factor 1 receptor (CSF1R) in PVMs was upregulated and activated in chronically SIV-infected rhesus macaques with encephalitis, correlating with SIV infection of PVMs. Herein, we investigated the role of CSF1R in the brain during acute SIV infection using BLZ945, a brain-penetrant CSF1R kinase inhibitor. Apart from three uninfected historic controls, nine Indian rhesus macaques were infected acutely with SIVmac251 and divided into three groups (n = 3 each): an untreated control and two groups treated for 20-30 days with low- (10 mg/kg/day) or high- (30 mg/kg/day) dose BLZ945. With the high-dose BLZ945 treatment, there was a significant reduction in cells expressing CD163 and CD206 across all four brain areas examined, compared with the low-dose treatment and control groups. In 9 of 11 tested regions, tissue viral DNA (vDNA) loads were reduced by 95%-99% following at least one of the two doses, and even to undetectable levels in some instances. Decreased numbers of CD163+ and CD206+ cells correlated significantly with lower levels of vDNA in all four corresponding brain areas. In contrast, BLZ945 treatment did not significantly affect the number of microglia. Our results indicate that doses as low as 10 mg/kg/day of BLZ945 are sufficient to reduce the tissue vDNA loads in the brain with no apparent adverse effect. This study provides evidence that infected PVMs are highly sensitive to CSF1R inhibition, opening new possibilities to achieve viral clearance.


Asunto(s)
Encéfalo , Macaca mulatta , Macrófagos , Síndrome de Inmunodeficiencia Adquirida del Simio , Virus de la Inmunodeficiencia de los Simios , Animales , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/virología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Carga Viral/efectos de los fármacos , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Antígenos CD/metabolismo , Masculino , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/virología , Antígenos de Diferenciación Mielomonocítica/metabolismo , Receptores de Superficie Celular/metabolismo , Anisoles
5.
J Infect Dis ; 230(2): 467-479, 2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-38135285

RESUMEN

Respiratory syncytial virus (RSV) primarily infects the respiratory epithelium, but growing evidence suggests that it may also be responsible for neurologic sequelae. In 3-dimensional microphysiologic peripheral nerve cultures, RSV infected neurons, macrophages, and dendritic cells along 2 distinct trajectories depending on the initial viral load. Low-level infection was transient, primarily involved macrophages, and induced moderate chemokine release with transient neural hypersensitivity. Infection with higher viral loads was persistent, infected neuronal cells in addition to monocytes, and induced robust chemokine release followed by progressive neurotoxicity. In spinal cord cultures, RSV infected microglia and dendritic cells but not neurons, producing a moderate chemokine expression pattern. The persistence of infection was variable but could be identified in dendritic cells as long as 30 days postinoculation. This study suggests that RSV can disrupt neuronal function directly through infection of peripheral neurons and indirectly through infection of resident monocytes and that inflammatory chemokines likely mediate both mechanisms.


Asunto(s)
Quimiocinas , Infecciones por Virus Sincitial Respiratorio , Médula Espinal , Quimiocinas/metabolismo , Médula Espinal/virología , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/virología , Animales , Neuronas/virología , Neuronas/metabolismo , Humanos , Nervios Periféricos/virología , Macrófagos/virología , Macrófagos/inmunología , Macrófagos/metabolismo , Carga Viral , Células Dendríticas/virología , Células Dendríticas/inmunología , Monocitos/virología , Monocitos/inmunología , Monocitos/metabolismo , Células Cultivadas , Virus Sincitiales Respiratorios/inmunología , Microglía/virología , Microglía/metabolismo
6.
Anal Chem ; 96(19): 7479-7486, 2024 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-38689560

RESUMEN

In the pathogenesis of microglia, brain immune cells promote nitrergic stress by overproducing nitric oxide (NO), leading to neuroinflammation. Furthermore, NO has been linked to COVID-19 progression, which has caused significant morbidity and mortality. SARS-CoV-2 infection activates inflammation by releasing excess NO and causing cell death in human microglial clone 3 (HMC3). In addition, NO regulates lysosomal functions and complex machinery to neutralize pathogens through phagocytosis. Therefore, developing lysosome-specific NO probes to monitor phagocytosis in microglia during the COVID-19 infection would be a significant study. Herein, a unique synthetic strategy was adopted to develop a NO selective fluorescent probe, PDM-NO, which can discriminate activated microglia from their resting state. The nonfluorescent PDM-NO exhibits a turn-on response toward NO only at lysosomal pH (4.5-5.5). Quantum chemical calculations (DFT/TD-DFT/PCM) and photophysical study revealed that the photoinduced electron transfer (PET) process is pivotal in tuning optical properties. PDM-NO demonstrated good biocompatibility and lysosomal specificity in activated HMC3 cells. Moreover, it can effectively map the dynamics of lysosomal NO against SARS-CoV-2 RNA-induced neuroinflammation in HMC3. Thus, PDM-NO is a potential fluorescent marker for detecting RNA virus infection and monitoring phagocytosis in HMC3.


Asunto(s)
COVID-19 , Colorantes Fluorescentes , Lisosomas , Microglía , Óxido Nítrico , Fagocitosis , SARS-CoV-2 , Microglía/virología , Microglía/metabolismo , SARS-CoV-2/aislamiento & purificación , Humanos , Lisosomas/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico/análisis , COVID-19/virología , COVID-19/diagnóstico , COVID-19/metabolismo , Colorantes Fluorescentes/química , ARN Viral/análisis , ARN Viral/metabolismo , Enfermedades Neuroinflamatorias , Línea Celular , Fenotipo
7.
J Neuroinflammation ; 21(1): 264, 2024 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-39425188

RESUMEN

Viral encephalitis is characterized by a series of immunological reactions that can control virus infection in the brain, but dysregulated responses may cause excessive inflammation and brain damage. Microglia are brain-resident myeloid cells that are specialized in surveilling the local CNS environment and in case of viral brain infection they contribute to the control of the infection and to restriction of viral dissemination. Here, we report that after exposure to neurotropic vesicular stomatitis virus (VSV), murine in vitro microglia cultures showed rapid upregulation of a broad range of pro-inflammatory and antiviral genes, which were stably expressed over the entire 8 h infection period. Additionally, a set of immunomodulatory genes was upregulated between 6 and 8 h post infection. In microglia cultures, the induction of several immune response pathways including cytokine responses was dependent on mitochondrial antiviral-signaling protein (MAVS). Consequently, in Mavs-deficient microglia the control of virus propagation failed as indicated by augmented virus titers and the accumulation of viral transcripts. Thus, in the analyzed in vitro system, MAVS signaling is critically required to achieve full microglia activation and to mediate profound antiviral effects. In Mavs-deficient mice, intranasal VSV instillation caused higher disease severity than in WT mice and virus dissemination was noticed beyond the olfactory bulb. Virus spread to inner regions of the olfactory bulb, i.e., the granular cell layer, correlated with the recruitment of highly inflammatory non-microglia myeloid cells into the olfactory bulb in Mavs-/- mice. Furthermore, increased cytokine levels were detected in the nasal cavity, the olfactory bulb and in other brain regions. Thus, microglial MAVS signaling is critically needed for virus sensing, full microglia activation, and for orchestration of protective immunity in the virus-infected CNS.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía , Transducción de Señal , Animales , Microglía/metabolismo , Microglía/virología , Microglía/inmunología , Ratones , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Células Cultivadas , Encefalitis Viral/inmunología , Encefalitis Viral/virología , Vesiculovirus/inmunología
8.
J Neuroinflammation ; 21(1): 203, 2024 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-39153993

RESUMEN

Microglia serve as a front-line defense against neuroinvasive viral infection, however, determination of their actual transcriptional profiles under conditions of health and disease is challenging. Here, we used various experimental approaches to delineate the transcriptional landscape of microglia during viral infection. Intriguingly, multiple activation genes were found to be artificially induced in sorted microglia and we demonstrated that shear stress encountered during cell sorting was one of the key inducers. Post-hoc analysis revealed that publicly available large-scale single-cell RNA sequencing datasets were significantly tainted by aberrant signatures that are associated with cell sorting. By exploiting the ribosomal tagging approach, we developed a strategy to enrich microglia-specific transcripts by comparing immunoprecipitated RNA with total RNA. Such enriched transcripts were instrumental in defining bona fide signatures of microglia under conditions of health and virus infection. These unified microglial signatures may serve as a benchmark to retrospectively assess ex vivo artefacts from available atlases. Leveraging the microglial translatome, we found enrichment of genes implicated in T-cell activation and cytokine production during the course of VSV infection. These data linked microglia with T-cell re-stimulation and further underscored that microglia are involved in shaping antiviral T-cell responses in the brain. Collectively, our study defines the transcriptional landscape of microglia under steady state and during viral encephalitis and highlights cellular interactions between microglia and T cells that contribute to the control of virus dissemination.


Asunto(s)
Encefalitis Viral , Perfilación de la Expresión Génica , Homeostasis , Microglía , Microglía/metabolismo , Microglía/virología , Animales , Encefalitis Viral/genética , Encefalitis Viral/inmunología , Encefalitis Viral/virología , Homeostasis/fisiología , Homeostasis/genética , Ratones , Perfilación de la Expresión Génica/métodos , Transcriptoma , Ratones Endogámicos C57BL
9.
J Neuroinflammation ; 21(1): 163, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38918792

RESUMEN

BACKGROUND: The SARS-CoV-2 virus activates maternal and placental immune responses. Such activation in the setting of other infections during pregnancy is known to impact fetal brain development. The effects of maternal immune activation on neurodevelopment are mediated at least in part by fetal brain microglia. However, microglia are inaccessible for direct analysis, and there are no validated non-invasive surrogate models to evaluate in utero microglial priming and function. We have previously demonstrated shared transcriptional programs between microglia and Hofbauer cells (HBCs, or fetal placental macrophages) in mouse models. METHODS AND RESULTS: We assessed the impact of maternal SARS-CoV-2 on HBCs isolated from 24 term placentas (N = 10 SARS-CoV-2 positive cases, 14 negative controls). Using single-cell RNA-sequencing, we demonstrated that HBC subpopulations exhibit distinct cellular programs, with specific subpopulations differentially impacted by SARS-CoV-2. Assessment of differentially expressed genes implied impaired phagocytosis, a key function of both HBCs and microglia, in some subclusters. Leveraging previously validated models of microglial synaptic pruning, we showed that HBCs isolated from placentas of SARS-CoV-2 positive pregnancies can be transdifferentiated into microglia-like cells (HBC-iMGs), with impaired synaptic pruning behavior compared to HBC models from negative controls. CONCLUSION: These findings suggest that HBCs isolated at birth can be used to create personalized cellular models of offspring microglial programming.


Asunto(s)
COVID-19 , Macrófagos , Microglía , Placenta , Complicaciones Infecciosas del Embarazo , SARS-CoV-2 , Femenino , Embarazo , Microglía/virología , Humanos , Placenta/virología , COVID-19/inmunología , Macrófagos/virología , Complicaciones Infecciosas del Embarazo/virología , Complicaciones Infecciosas del Embarazo/patología , SARS-CoV-2/patogenicidad , Feto , Adulto , Encéfalo/virología , Encéfalo/patología , Ratones , Animales
10.
J Neuroinflammation ; 21(1): 176, 2024 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-39026249

RESUMEN

Accumulating evidence implicates that herpes simplex virus type 1 (HSV-1) has been linked to the development and progression of Alzheimer's disease (AD). HSV-1 infection induces ß-amyloid (Aß) deposition in vitro and in vivo, but the effect and precise mechanism remain elusive. Here, we show that HSV-1 infection of the brains of transgenic 5xFAD mice resulted in accelerated Aß deposition, gliosis, and cognitive dysfunction. We demonstrate that HSV-1 infection induced the recruitment of microglia to the viral core to trigger microglial phagocytosis of HSV-GFP-positive neuronal cells. In addition, we reveal that the NLRP3 inflammasome pathway induced by HSV-1 infection played a crucial role in Aß deposition and the progression of AD caused by HSV-1 infection. Blockade of the NLRP3 inflammasome signaling reduces Aß deposition and alleviates cognitive decline in 5xFAD mice after HSV-1 infection. Our findings support the notion that HSV-1 infection is a key factor in the etiology of AD, demonstrating that NLRP3 inflammasome activation functions in the interface of HSV-1 infection and Aß deposition in AD.


Asunto(s)
Enfermedad de Alzheimer , Progresión de la Enfermedad , Herpesvirus Humano 1 , Ratones Transgénicos , Microglía , Proteína con Dominio Pirina 3 de la Familia NLR , Fagocitosis , Transducción de Señal , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/virología , Ratones , Microglía/metabolismo , Microglía/patología , Microglía/virología , Transducción de Señal/fisiología , Humanos , Herpes Simple/patología , Herpes Simple/inmunología , Herpes Simple/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Modelos Animales de Enfermedad , Presenilina-1/genética , Presenilina-1/metabolismo
11.
J Virol ; 97(12): e0159523, 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-38032195

RESUMEN

IMPORTANCE: Our mouse model is a powerful tool for investigating the genetic mechanisms governing central nervous system (CNS) human immunodeficiency virus type-1 (HIV-1) infection and latency in the CNS at a single-cell level. A major advantage of our model is that it uses induced pluripotent stem cell-derived microglia, which enables human genetics, including gene function and therapeutic gene manipulation, to be explored in vivo, which is more challenging to study with current hematopoietic stem cell-based models for neuroHIV. Our transgenic tracing of xenografted human cells will provide a quantitative medium to develop new molecular and epigenetic strategies for reducing the HIV-1 latent reservoir and to test the impact of therapeutic inflammation-targeting drug interventions on CNS HIV-1 latency.


Asunto(s)
Infecciones por VIH , VIH-1 , Células Madre Pluripotentes Inducidas , Microglía , Animales , Humanos , Ratones , Sistema Nervioso Central , Infecciones por VIH/metabolismo , Infecciones por VIH/patología , VIH-1/fisiología , Microglía/virología , Latencia del Virus , Xenoinjertos
12.
PLoS Pathog ; 18(7): e1010110, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35797416

RESUMEN

Human immune deficiency virus (HIV) infection in the brain leads to chronic neuroinflammation due to the production of pro-inflammatory cytokines, which in turn promotes HIV transcription in infected microglial cells. However, powerful counteracting silencing mechanisms in microglial cells result in the rapid shutdown of HIV expression after viral reactivation to limit neuronal damage. Here we investigated whether the Nerve Growth Factor IB-like nuclear receptor Nurr1 (NR4A2), which is a repressor of inflammation in the brain, acts directly to restrict HIV expression. HIV silencing following activation by TNF-α, or a variety of toll-like receptor (TLR) agonists, in both immortalized human microglial cells (hµglia) and induced pluripotent stem cells (iPSC)-derived human microglial cells (iMG) was enhanced by Nurr1 agonists. Similarly, overexpression of Nurr1 led to viral suppression, while conversely, knock down (KD) of endogenous Nurr1 blocked HIV silencing. The effect of Nurr1 on HIV silencing is direct: Nurr1 binds directly to the specific consensus binding sites in the U3 region of the HIV LTR and mutation of the Nurr1 DNA binding domain blocked its ability to suppress HIV-1 transcription. Chromatin immunoprecipitation (ChIP) assays also showed that after Nurr1 binding to the LTR, the CoREST/HDAC1/G9a/EZH2 transcription repressor complex is recruited to the HIV provirus. Finally, transcriptomic studies demonstrated that in addition to repressing HIV transcription, Nurr1 also downregulated numerous cellular genes involved in inflammation, cell cycle, and metabolism, further promoting HIV latency and microglial homoeostasis. Nurr1 therefore plays a pivotal role in modulating the cycles of proviral reactivation by potentiating the subsequent proviral transcriptional shutdown. These data highlight the therapeutic potential of Nurr1 agonists for inducing HIV silencing and microglial homeostasis and ultimately for the amelioration of the neuroinflammation associated with HIV-associated neurocognitive disorders (HAND).


Asunto(s)
Infecciones por VIH , VIH-1 , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Humanos , Inflamación/metabolismo , Microglía/metabolismo , Microglía/virología , Factores de Crecimiento Nervioso/metabolismo , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Provirus
13.
J Med Virol ; 96(10): e29945, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39370874

RESUMEN

Borna disease virus 1 (BoDV-1) is a neurotropic RNA virus that has been linked to fatal BoDV-1 encephalitis (BVE) in humans. Ferroptosis represents a newly recognized kind of programmed cell death that marked by iron overload and lipid peroxidation. Various viral infections are closely related to ferroptosis. However, the link between BoDV-1 infection and ferroptosis, as well as its role in BVE pathogenesis, remains inadequately understood. Herein, we used primary rat cortical neurons, human microglial HMC3 cells, and Sprague‒Dawley rats as models. BoDV-1 infection induced ferroptosis, as ferroptosis characteristics were detected (iron overload, reactive oxygen species buildup, decreased antioxidant capacity, lipid peroxidation, and mitochondrial damage). Analysis via qRT-PCR and Western blot demonstrated that BoDV-1-induced ferroptosis was mediated through Nrf2/HO-1/SLC7a11/GPX4 antioxidant pathway suppression. Nrf2 downregulation was due to BoDV-1 infection promoting Nrf2 ubiquitination and degradation. Following BoDV-1-induced ferroptosis, the PTGS2/PGE2 signaling pathway was activated, and various intracellular lipid peroxidation products and damage-associated molecular patterns were released, contributing to BVE occurrence and progression. More importantly, inhibiting ferroptosis or the ubiquitin‒proteasome system effectively alleviated BVE. Collectively, these findings demonstrate the interaction between BoDV-1 infection and ferroptosis and reveal BoDV-1-induced ferroptosis as an underlying pathogenic mechanism of BVE.


Asunto(s)
Enfermedad de Borna , Virus de la Enfermedad de Borna , Ferroptosis , Peroxidación de Lípido , Factor 2 Relacionado con NF-E2 , Neuronas , Ratas Sprague-Dawley , Virus de la Enfermedad de Borna/fisiología , Animales , Ratas , Humanos , Neuronas/virología , Neuronas/patología , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/genética , Enfermedad de Borna/virología , Enfermedad de Borna/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Sistema de Transporte de Aminoácidos y+/metabolismo , Sistema de Transporte de Aminoácidos y+/genética , Ciclooxigenasa 2/metabolismo , Ciclooxigenasa 2/genética , Microglía/virología , Microglía/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Hemo-Oxigenasa 1/metabolismo , Hemo-Oxigenasa 1/genética , Línea Celular , Encefalitis/virología , Encefalitis/patología , Células Cultivadas
14.
J Neurovirol ; 30(3): 251-265, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38842651

RESUMEN

Japanese Encephalitis remains a significant global health concern, contributing to millions of deaths annually worldwide. Microglial cells, as key innate immune cells within the central nervous system (CNS), exhibit intricate cellular structures and possess molecular phenotypic plasticity, playing pivotal roles in immune responses during CNS viral infections. Particularly under viral inflammatory conditions, microglial cells orchestrate innate and adaptive immune responses to mitigate viral invasion and dampen inflammatory reactions. This review article comprehensively summarizes the pathophysiology of viral invasion into the CNS and the cellular interactions involved, elucidating the roles of various immune mediators, including pro-inflammatory cytokines, in neuroinflammation. Leveraging this knowledge, strategies for modulating inflammatory responses and attenuating hyperactivation of glial cells to mitigate viral replication within the brain are discussed. Furthermore, current chemotherapeutic and antiviral drugs are examined, elucidating their mechanisms of action against viral replication. This review aims to provide insights into therapeutic interventions for Japanese Encephalitis and related viral infections, ultimately contributing to improved outcomes for affected individuals.


Asunto(s)
Citocinas , Virus de la Encefalitis Japonesa (Especie) , Encefalitis Japonesa , Microglía , Humanos , Encefalitis Japonesa/inmunología , Encefalitis Japonesa/virología , Encefalitis Japonesa/patología , Encefalitis Japonesa/tratamiento farmacológico , Encefalitis Japonesa/terapia , Virus de la Encefalitis Japonesa (Especie)/inmunología , Virus de la Encefalitis Japonesa (Especie)/patogenicidad , Citocinas/inmunología , Microglía/inmunología , Microglía/virología , Microglía/patología , Animales , Antivirales/uso terapéutico , Replicación Viral/inmunología , Inmunidad Innata , Inmunoterapia/métodos , Encéfalo/virología , Encéfalo/inmunología , Encéfalo/patología , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/virología , Enfermedades Neuroinflamatorias/patología , Enfermedades Neuroinflamatorias/tratamiento farmacológico
15.
J Neurovirol ; 30(4): 362-379, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38600307

RESUMEN

The human immunodeficiency virus (HIV) epidemic is an ongoing global health problem affecting 38 million people worldwide with nearly 1.6 million new infections every year. Despite the advent of combined antiretroviral therapy (cART), a large percentage of people with HIV (PWH) still develop neurological deficits, grouped into the term of HIV-associated neurocognitive disorders (HAND). Investigating the neuropathology of HIV is important for understanding mechanisms associated with cognitive impairment seen in PWH. The major obstacle for studying neuroHIV is the lack of suitable in vitro human culture models that could shed light into the HIV-CNS interactions. Recent advances in induced pluripotent stem cell (iPSC) culture and 3D brain organoid systems have allowed the generation of 2D and 3D culture methods that possess a potential to serve as a model of neurotropic viral diseases, including HIV. In this study, we first generated and characterized several hiPSC lines from healthy human donor skin fibroblast cells. hiPSCs were then used for the generation of microglia-containing human cerebral organoids (hCOs). Once fully characterized, hCOs were infected with HIV-1 in the presence and absence of cART regimens and viral infection was studied by cellular, molecular/biochemical, and virological assays. Our results revealed that hCOs were productively infected with HIV-1 as evident by viral p24-ELISA in culture media, RT-qPCR and RNAscope analysis of viral RNA, as well as ddPCR analysis of proviral HIV-1 in genomic DNA samples. More interestingly, replication and gene expression of HIV-1 were also greatly suppressed by cART in hCOs as early as 7 days post-infections. Our results suggest that hCOs derived from hiPSCs support HIV-1 replication and gene expression and may serve as a unique platform to better understand neuropathology of HIV infection in the brain.


Asunto(s)
VIH-1 , Células Madre Pluripotentes Inducidas , Microglía , Organoides , Humanos , Células Madre Pluripotentes Inducidas/virología , VIH-1/fisiología , VIH-1/genética , VIH-1/patogenicidad , Organoides/virología , Microglía/virología , Microglía/patología , Replicación Viral/efectos de los fármacos , Infecciones por VIH/virología , Infecciones por VIH/patología , Infecciones por VIH/tratamiento farmacológico , Encéfalo/virología , Encéfalo/patología , Complejo SIDA Demencia/virología , Complejo SIDA Demencia/patología
16.
J Neurovirol ; 30(4): 380-392, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38713307

RESUMEN

Despite antiretroviral therapy (ART), HIV persistence in the central nervous system (CNS) continues to cause a range of cognitive impairments in people living with HIV (PLWH). Upon disease progression, transmigrating CCR5-using T-cell tropic viruses are hypothesized to evolve into macrophage-tropic viruses in the CNS that can efficiently infect low CD4-expressing cells, such as microglia. We examined HIV-1 RNA concentration, co-receptor usage, and CSF compartmentalization in paired CSF and blood samples from 19 adults not on treatment. Full-length envelope CSF- and plasma-derived reporter viruses were generated from 3 subjects and phenotypically characterized in human primary CD4+ T-cells and primary microglia. Median HIV RNA levels were higher in plasma than in CSF (5.01 vs. 4.12 log10 cp/mL; p = 0.004), and coreceptor usage was mostly concordant for CCR5 across the paired samples (n = 17). Genetically compartmentalized CSF viral populations were detected in 2 subjects, one with and one without neurological symptoms. All viral clones could replicate in T-cells (R5 T cell-tropic). In addition, 3 CSF and 1 plasma patient-derived viral clones also had the capacity to replicate in microglia/macrophages and, therefore have an intermediate macrophage tropic phenotype. Overall, with this study, we demonstrate that in a subset of PLWH, plasma-derived viruses undergo genetic and phenotypic evolution within the CNS, indicating viral infection and replication in CNS cells. It remains to be studied whether the intermediate macrophage-tropic phenotype observed in primary microglia represents a midpoint in the evolution towards a macrophage-tropic phenotype that can efficiently replicate in microglial cells and propagate viral infection in the CNS.


Asunto(s)
Linfocitos T CD4-Positivos , Infecciones por VIH , VIH-1 , Microglía , ARN Viral , Receptores CCR5 , Replicación Viral , Humanos , Microglía/virología , Microglía/patología , Microglía/inmunología , VIH-1/patogenicidad , VIH-1/genética , Linfocitos T CD4-Positivos/virología , Linfocitos T CD4-Positivos/inmunología , Receptores CCR5/genética , Infecciones por VIH/líquido cefalorraquídeo , Infecciones por VIH/virología , Infecciones por VIH/sangre , Infecciones por VIH/inmunología , Adulto , Masculino , Femenino , ARN Viral/líquido cefalorraquídeo , ARN Viral/sangre , ARN Viral/genética , Persona de Mediana Edad , Tropismo Viral , Carga Viral , Receptores CXCR4/genética , Células Cultivadas
17.
J Neurovirol ; 30(3): 215-228, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38922550

RESUMEN

The cellular prion protein (PrPC) is an extracellular cell membrane protein. Due to its diversified roles, a definite role of PrPC has been difficult to establish. During viral infection, PrPC has been reported to play a pleiotropic role. Here, we have attempted to envision the function of PrPC in the neurotropic m-CoV-MHV-RSA59-induced model of neuroinflammation in C57BL/6 mice. A significant upregulation of PrPC at protein and mRNA levels was evident in infected mouse brains during the acute phase of neuroinflammation. Furthermore, investigation of the effect of MHV-RSA59 infection on PrPC expression in specific neuronal, microglial, and astrocytoma cell lines, revealed a differential expression of prion protein during neuroinflammation. Additionally, siRNA-mediated downregulation of prnp transcripts reduced the expression of viral antigen and viral infectivity in these cell lines. Cumulatively, our results suggest that PrPC expression significantly increases during acute MHV-RSA59 infection and that PrPC also assists in viral infectivity and viral replication.


Asunto(s)
Ratones Endogámicos C57BL , Microglía , Virus de la Hepatitis Murina , Enfermedades Neuroinflamatorias , Proteínas PrPC , Animales , Virus de la Hepatitis Murina/patogenicidad , Ratones , Proteínas PrPC/metabolismo , Proteínas PrPC/genética , Enfermedades Neuroinflamatorias/virología , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/genética , Enfermedades Neuroinflamatorias/patología , Microglía/metabolismo , Microglía/virología , Microglía/patología , Encéfalo/virología , Encéfalo/metabolismo , Encéfalo/patología , Neuronas/virología , Neuronas/metabolismo , Neuronas/patología , Replicación Viral , Línea Celular Tumoral , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Regulación hacia Arriba , Línea Celular , Humanos , Modelos Animales de Enfermedad , Proteínas Priónicas
18.
J Neurovirol ; 30(4): 337-352, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38884890

RESUMEN

HIV-associated neurological disorder (HAND) is a serious complication of HIV infection marked by neurotoxicity induced by viral proteins like Tat. Substance abuse exacerbates neurocognitive impairment in people living with HIV. There is an urgent need for therapeutic strategies to combat HAND comorbid with Cocaine Use Disorder (CUD). Our analysis of HIV and cocaine-induced transcriptomes in primary cortical cultures revealed significant overexpression of the macrophage-specific gene aconitate decarboxylase 1 (Acod1). The ACOD1 protein converts the tricarboxylic acid intermediate cis-aconitate into itaconate during the activation of inflammation. Itaconate then facilitates cytokine production and activates anti-inflammatory transcription factors, shielding macrophages from infection-induced cell death. However, the immunometabolic function of itaconate was unexplored in HIV and cocaine-exposed microglia. We assessed the potential of 4-octyl-itaconate (4OI), a cell-penetrable ester form of itaconate known for its anti-inflammatory properties. When primary cortical cultures exposed to Tat and cocaine were treated with 4OI, microglial cell number increased and the morphological altercations induced by Tat and cocaine were reversed. Microglial cells also appeared more ramified, resembling the quiescent microglia. 4OI treatment inhibited secretion of the proinflammatory cytokines IL-1α, IL-1ß, IL-6, and MIP1-α induced by Tat and cocaine. Transcriptome profiling determined that Nrf2 target genes were significantly activated in Tat and 4OI treated cultures relative to Tat alone. Further, genes associated with cytoskeleton dynamics in inflammatory microglia were downregulated by 4OI treatment. Together, the results strongly suggest 4-octyl-itaconate holds promise as a potential candidate for therapeutic development to treat HAND coupled with CUD comorbidities.


Asunto(s)
Carboxiliasas , Cocaína , Microglía , Factor 2 Relacionado con NF-E2 , Succinatos , Productos del Gen tat del Virus de la Inmunodeficiencia Humana , Succinatos/farmacología , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , Microglía/virología , Animales , Cocaína/farmacología , Cocaína/efectos adversos , Carboxiliasas/genética , Carboxiliasas/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Humanos , Inflamación/tratamiento farmacológico , Inflamación/patología , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/virología , Infecciones por VIH/patología , Células Cultivadas , Trastornos Relacionados con Cocaína/tratamiento farmacológico , Trastornos Relacionados con Cocaína/metabolismo , Trastornos Relacionados con Cocaína/genética , Citocinas/genética , Citocinas/metabolismo , Ratas , Síndromes de Neurotoxicidad/tratamiento farmacológico , Síndromes de Neurotoxicidad/patología , Síndromes de Neurotoxicidad/metabolismo , Síndromes de Neurotoxicidad/genética , Cultivo Primario de Células , Regulación de la Expresión Génica/efectos de los fármacos
19.
Virol J ; 21(1): 165, 2024 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-39075520

RESUMEN

Echovirus 30 (E30), a member of the species B Enterovirus family, is a primary pathogen responsible for aseptic meningitis and encephalitis. E30 is associated with severe nervous system diseases and is a primary cause of child illness, disability, and even mortality. However, the mechanisms underlying E30-induced brain injury remain poorly understood. In this study, we used a neonatal mouse model of E30 to investigate the possible mechanisms of brain injury. E30 infection triggered the activation of microglia in the mouse brain and efficiently replicated within HMC3 cells. Subsequent transcriptomic analysis revealed inflammatory activation of microglia in response to E30 infection. We also detected a significant upregulation of polo-like kinase 1 (PLK1) and found that its inhibition could limit E30 infection in a sucking mouse model. Collectively, E30 infection led to brain injury in a neonatal mouse model, which may be related to excessive inflammatory responses. Our findings highlight the intricate interplay between E30 infection and neurological damage, providing crucial insights that could guide the development of interventions and strategies to address the severe clinical manifestations associated with this pathogen.


Asunto(s)
Encéfalo , Modelos Animales de Enfermedad , Enterovirus Humano B , Microglía , Animales , Ratones , Encéfalo/virología , Encéfalo/patología , Enterovirus Humano B/patogenicidad , Enterovirus Humano B/fisiología , Microglía/virología , Microglía/inmunología , Línea Celular , Humanos , Inflamación/virología , Animales Lactantes , Animales Recién Nacidos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Infecciones por Echovirus/virología , Replicación Viral
20.
Virol J ; 21(1): 219, 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39285274

RESUMEN

BACKGROUND: To investigate the mechanism of Golgi matrix protein 130(GM130) regulating the antiviral immune response of TLR3 after herpes simplex virus type 1(HSV-1) infection of microglia cells. We explored the regulatory effects of berberine on the immune response mediated by GM130 and TLR3. METHODS: An in vitro model of HSV-1 infection was established by infecting BV2 cells with HSV-1. RESULTS: Compared to the uninfected group, the Golgi apparatus (GA) fragmentation and GM130 decreased after HSV-1 infection; TLR3 increased at 6 h and began to decrease at 12 h after HSV-1 infection; the secretion of interferon-beta(IFN-ß), tumour necrosis factor alpha(TNF-α), and interleukin-6(IL-6) increased after infection. Knockdown of GM130 aggravated fragmentation of the GA and caused TLR3 to further decrease, and the virus titer also increased significantly. GM130 knockdown inhibits the increase in TLR3 and inflammatory factors induced by TLR3 agonists and increases the viral titer. Overexpression of GM130 alleviated fragmentation of the GA induced by HSV-1, partially restored the levels of TLR3, and reduced viral titers. GM130 overexpression reversed the reduction in TLR3 and inflammatory cytokine levels induced by TLR3 inhibitors. Therefore, the decrease in GM130 levels caused by HSV-1 infection leads to increased viral replication by inhibiting TLR3-mediated innate immunity. Berberine can protect the GA and reverse the downregulation of GM130, as well as the downregulation of TLR3 and its downstream factors after HSV-1 infection, reducing the virus titer. CONCLUSIONS: In microglia, one mechanism of HSV-1 immune escape is disruption of the GM130/TLR3 pathway. Berberine protects the GA and enhances TLR3-mediated antiviral immune responses.


Asunto(s)
Regulación hacia Abajo , Herpesvirus Humano 1 , Inmunidad Innata , Microglía , Receptor Toll-Like 3 , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 1/fisiología , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 3/genética , Microglía/virología , Microglía/inmunología , Microglía/efectos de los fármacos , Animales , Ratones , Línea Celular , Evasión Inmune , Berberina/farmacología , Citocinas/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Herpes Simple/inmunología , Herpes Simple/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA