Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(7)2024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-38612400

RESUMEN

Human Immunodeficiency Virus type 1 (HIV-1)-associated neurocognitive disorders (HANDs) remain prevalent in HIV-1-infected individuals despite the evident success of combined antiretroviral therapy (cART). The mechanisms underlying HAND prevalence in the cART era remain perplexing. Ample evidence indicates that HIV-1 envelope glycoprotein protein 120 (gp120), a potent neurotoxin, plays a pivotal role in HAND pathogenesis. Methamphetamine (Meth) abuse exacerbates HANDs, but how this occurs is not fully understood. We hypothesize that Meth exacerbates HANDs by enhancing gp120-mediated neuroinflammation. To test this hypothesis, we studied the effect of Meth on gp120-induced microglial activation and the resultant production of proinflammatory cytokines in primary rat microglial cultures. Our results show that Meth enhanced gp120-induced microglial activation, as revealed by immunostaining and Iba-1 expression, and potentiated gp120-mediated NLRP3 expression and IL-1ß processing and release, as assayed by immunoblotting and ELISA. Meth also augmented the co-localization of NLRP3 and caspase-1, increased the numbers of NLRP3 puncta and ROS production, increased the levels of iNOS expression and NO production, and increased the levels of cleaved gasderminD (GSDMD-N; an executor of pyroptosis) in gp120-primed microglia. The Meth-associated effects were attenuated or blocked by MCC950, an NLRP3 inhibitor, or Mito-TEMPO, a mitochondrial superoxide scavenger. These results suggest that Meth enhances gp120-associated microglial NLRP3 activation and the resultant proinflammatory responses via mitochondria-dependent signaling.


Asunto(s)
Trastornos Relacionados con Anfetaminas , VIH-1 , Animales , Ratas , Glicoproteínas , Inflamasomas , Microglía , Proteína con Dominio Pirina 3 de la Familia NLR
2.
Neurobiol Dis ; 168: 105712, 2022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-35337950

RESUMEN

Methamphetamine (Meth) abuse and human immunodeficiency virus type 1 (HIV-1) infection are two major public health problems worldwide. Being frequently comorbid with HIV-1 infection, Meth abuse exacerbates neurocognitive impairment in HIV-1-infected individuals even in the era of combined antiretroviral therapy. While a large body of research have studied the individual effects of Meth and HIV-1 envelope glycoprotein 120 (gp120) in the brain, far less has focused on their synergistic influence. Moreover, it is well-documented that the hippocampus is the primary site of spatial learning and long-term memory formation. Dysregulation of activity-dependent synaptic transmission and plasticity in the hippocampus is believed to impair neurocognitive function. To uncover the underlying mechanisms for increased incidence and severity of HIV-1-associated neurocognitive disorders (HAND) in HIV-1-infected patients with Meth abuse, we investigated acute individual and combined effects of Meth (20 µM) and gp120 (200 pM) on synaptic transmission and plasticity in the CA1 region of young adult male rat hippocampus, a brain region known to be vulnerable to HIV-1 infection. Our results showed that acute localized application of Meth and gp120 each alone onto the CA1 region reduced short-term dynamics of input-output responses and frequency facilitation, and attenuated long-term potentiation (LTP) induced by either high frequency stimulation or theta burst stimulation. A synergistic augmentation on activity-dependent synaptic plasticity was observed when Meth and gp120 were applied in combination. Paired-pulse facilitation results exhibited an altered facilitation ratio, suggesting a presynaptic site of action. Further studies revealed an involvement of microglia NLRP3 inflammasome activation in Meth augmentation of gp120-mediated attenuation of LTP. Taken together, our results demonstrated Meth augmented gp120 attenuation of LTP in the hippocampus. Since LTP is the accepted experimental analog of learning at the synaptic level, such augmentation may underlie Meth exacerbation of HAND observed clinically.


Asunto(s)
Infecciones por VIH , VIH-1 , Metanfetamina , Animales , Glicoproteínas/farmacología , Infecciones por VIH/complicaciones , Hipocampo , Humanos , Potenciación a Largo Plazo/fisiología , Masculino , Metanfetamina/farmacología , Trastornos Neurocognitivos , Plasticidad Neuronal , Ratas , Transmisión Sináptica
3.
J Neuroinflammation ; 18(1): 100, 2021 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-33902641

RESUMEN

BACKGROUND: Microglia are resident innate immune cells in the brain, and activation of these myeloid cells results in secretion of a variety of pro-inflammatory molecules, leading to the development of neurodegenerative disorders. Lipopolysaccharide (LPS) is a widely used experimental stimulant in microglia activation. We have previously shown that LPS produced microglia activation and evoked detectable functional abnormalities in rat corpus callosum (CC) in vitro. Here, we further validated the effects of low-dose LPS-induced microglia activation and resultant white matter abnormality in the CC in an animal model and examined its attenuation by an anti-inflammatory agent minocycline. METHODS: Twenty-four SD rats were divided randomly into three groups and intra-peritoneally injected daily with saline, LPS, and LPS + minocycline, respectively. All animals were subject to MRI tests 6 days post-injection. The animals were then sacrificed to harvest the CC tissues for electrophysiology, western blotting, and immunocytochemistry. One-way ANOVA with Tukey's post-test of all pair of columns was employed statistical analyses. RESULTS: Systemic administration of LPS produced microglial activation in the CC as illustrated by Iba-1 immunofluorescent staining. We observed that a large number of Iba-1-positive microglial cells were hyper-ramified with hypertrophic somata or even amoeba like in the LPS-treated animals, and such changes were significantly reduced by co-administration of minocycline. Electrophysiological recordings of axonal compound action potential (CAP) in the brain slices contained the CC revealed an impairment on the CC functionality as detected by a reduction in CAP magnitude. Such an impairment was supported by a reduction of fast axonal transportation evidenced by ß-amyloid precursor protein accumulation. These alterations were attenuated by minocycline, demonstrating minocycline reduction of microglia-mediated interruption of white matter integrity and function in the CC. CONCLUSIONS: Systemic administration of LPS produced microglia activation in the CC and resultant functional abnormalities that were attenuated by an anti-inflammatory agent minocycline.


Asunto(s)
Cuerpo Calloso/patología , Microglía/patología , Minociclina/uso terapéutico , Animales , Antibacterianos/farmacología , Cuerpo Calloso/diagnóstico por imagen , Cuerpo Calloso/efectos de los fármacos , Cuerpo Calloso/fisiopatología , Lipopolisacáridos/farmacología , Imagen por Resonancia Magnética , Microglía/efectos de los fármacos , Microglía/inmunología , Microglía/metabolismo , Ratas , Ratas Sprague-Dawley , Sustancia Blanca/diagnóstico por imagen , Sustancia Blanca/metabolismo , Sustancia Blanca/patología
4.
Mol Cell Neurosci ; 82: 167-175, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28552341

RESUMEN

Methamphetamine (Meth) abuse not only increases the risk of human immunodeficiency virus-1 (HIV-1) infection, but exacerbates HIV-1-associated neurocognitive disorders (HAND) as well. The mechanisms underlying the co-morbid effect are not fully understood. Meth and HIV-1 each alone interacts with microglia and microglia express voltage-gated potassium (KV) channel KV1.3. To understand whether KV1.3 functions an intersecting point for Meth and HIV-1, we studied the augment effect of Meth on HIV-1 glycoprotein 120 (gp120)-induced neurotoxic activity in cultured rat microglial cells. While Meth and gp120 each alone at low (subtoxic) concentrations failed to trigger microglial neurotoxic activity, Meth potentiated gp120-induced microglial neurotoxicity when applied in combination. Meth enhances gp120 effect on microglia by enhancing microglial KV1.3 protein expression and KV1.3 current, leading to an increase of neurotoxin production and resultant neuronal injury. Pretreatment of microglia with a specific KV1.3 antagonist 5-(4-Phenoxybutoxy)psoralen (PAP) or a broad spectrum KV channel blocker 4-aminopyridine (4-AP) significantly attenuated Meth/gp120-treated microglial production of neurotoxins and resultant neuronal injury, indicating an involvement of KV1.3 in Meth/gp120-induced microglial neurotoxic activity. Meth/gp120 activated caspase-3 and increased caspase-3/7 activity in microglia and inhibition of caspase-3 by its specific inhibitor significantly decreased microglial production of TNF-α and iNOS and attenuated microglia-associated neurotoxic activity. Moreover, blockage of KV1.3 by specific blockers attenuated Meth/gp120 enhancement of caspase-3/7 activity. Taking together, these results suggest an involvement of microglial KV1.3 in the mediation of Meth/gp120 co-morbid effect on microglial neurotoxic activity via caspase-3 signaling.


Asunto(s)
Proteína gp120 de Envoltorio del VIH/metabolismo , Metanfetamina/farmacología , Microglía/metabolismo , Potasio/metabolismo , Animales , Células Cultivadas , Femenino , Neuronas/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
5.
Neurobiol Dis ; 97(Pt A): 1-10, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27816768

RESUMEN

Brain white matter damage is frequently detected in patients infected with human immunodeficiency virus type 1 (HIV-1). White matter is composed of neuronal axons sheathed by oligodendrocytes (Ols), the myelin-forming cells in central nervous system. Ols are susceptible to HIV-1 viral trans-activator of transcription (Tat) and injury of Ols results in myelin sheath damage. It has been demonstrated that activation of voltage-gated K+ (KV) channels induces cell apoptosis and Ols predominantly express K+ channel KV1.3. It is our hypothesis that Tat injures Ols via activation of KV1.3. To test this hypothesis, we studied the involvement of KV1.3 in Tat-induced Ol/myelin injury both in vitro and ex vivo. Application of Tat to primary rat Ol cultures enhanced whole-cell KV1.3 current recorded under voltage clamp configuration and confirmed by specific KV1.3 antagonists Margatoxin (MgTx) and 5-(4-phenoxybutoxy) psoralen (PAP). The Tat enhancement of KV1.3 current was associated with Tat-induced Ol apoptosis, which was blocked by MgTx and PAP or by siRNA knockdown of KV1.3 gene. The Tat-induced Ol injury was validated in cultured rat brain slices, particularly in corpus callosum and striatum, that incubation of the slices with Tat resulted in myelin damage and reduction of myelin basic protein which were also blocked by aforementioned KV1.3 antagonists. Further studies revealed that Tat interacts with KV1.3 as determined by protein pull-down of recombinant GST-Tat with KV1.3 expressed in rat brains and HEK293 cells. Such protein-protein interaction may alter channel protein phosphorylation, resultant channel activity and consequent Ol/myelin injury. Taken together, these results demonstrate an involvement of KV1.3 in Tat- induced Ol/myelin injury, a potential mechanism for the pathogenesis of HIV-1-associated white matter damage.


Asunto(s)
Canal de Potasio Kv1.3/metabolismo , Oligodendroglía/metabolismo , Potasio/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Cationes Monovalentes/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células HEK293 , VIH-1 , Humanos , Canal de Potasio Kv1.3/antagonistas & inhibidores , Canal de Potasio Kv1.3/genética , Proteína Básica de Mielina/metabolismo , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/metabolismo , Vaina de Mielina/patología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/patología , Oligodendroglía/virología , Fosforilación , Ratas Sprague-Dawley , Técnicas de Cultivo de Tejidos
6.
Mol Cell Neurosci ; 57: 73-82, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24416794

RESUMEN

Plasma gelsolin (pGSN), a secreted form of gelsolin, is constitutively expressed throughout the central nervous system (CNS). The neurons, astrocytes and oligodendrocytes are the major sources of pGSN in the CNS. It has been shown that levels of pGSN in the cerebrospinal fluid (CSF) are decreased in several neurological conditions including HIV-1-associated neurocognitive disorders (HAND). Although there is no direct evidence that a decreased level of pGSN in CSF is causally related to the pathogenesis of neurological disorders, neural cells, if lacking pGSN, are more vulnerable to cell death. To understand how GSN levels relate to neuronal injury in HAND, we studied the effects of pGSN on HIV-1 gp120-activated outward K+ currents in primary rat cortical neuronal cultures. Incubation of rat cortical neurons with gp120 enhanced the outward K+ currents induced by voltage steps and resulted in neuronal apoptosis. Treatment with pGSN suppressed the gp120-induced increase of delayed rectifier current (IK) and reduced vulnerability to gp120-induced neuronal apoptosis. Application of Guangxitoxin-1E (GxTx), a Kv2.1 specific channel inhibitor, inhibited gp120 enhancement of IK and associated neuronal apoptosis, similar effects to pGSN. Western blot and PCR analysis revealed gp120 exposure to up-regulate Kv2.1 channel expression, which was also inhibited by treatment with pGSN. Taken together, these results indicate pGSN protects neurons by suppressing gp120 enhancement of IK through Kv2.1 channels and reduction of pGSN in HIV-1-infected brain may contribute to HIV-1-associated neuropathy.


Asunto(s)
Apoptosis , Gelsolina/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Canales de Potasio Shab/metabolismo , Potenciales de Acción , Animales , Células Cultivadas , Corteza Cerebral/citología , Proteína gp120 de Envoltorio del VIH/toxicidad , Neuronas/metabolismo , Neuronas/patología , Neuronas/fisiología , Bloqueadores de los Canales de Potasio/farmacología , Ratas , Ratas Sprague-Dawley , Canales de Potasio Shab/antagonistas & inhibidores
7.
Bioengineering (Basel) ; 11(6)2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38927781

RESUMEN

Automatically segmenting polyps from colonoscopy videos is crucial for developing computer-assisted diagnostic systems for colorectal cancer. Existing automatic polyp segmentation methods often struggle to fulfill the real-time demands of clinical applications due to their substantial parameter count and computational load, especially those based on Transformer architectures. To tackle these challenges, a novel lightweight long-range context fusion network, named LightCF-Net, is proposed in this paper. This network attempts to model long-range spatial dependencies while maintaining real-time performance, to better distinguish polyps from background noise and thus improve segmentation accuracy. A novel Fusion Attention Encoder (FAEncoder) is designed in the proposed network, which integrates Large Kernel Attention (LKA) and channel attention mechanisms to extract deep representational features of polyps and unearth long-range dependencies. Furthermore, a newly designed Visual Attention Mamba module (VAM) is added to the skip connections, modeling long-range context dependencies in the encoder-extracted features and reducing background noise interference through the attention mechanism. Finally, a Pyramid Split Attention module (PSA) is used in the bottleneck layer to extract richer multi-scale contextual features. The proposed method was thoroughly evaluated on four renowned polyp segmentation datasets: Kvasir-SEG, CVC-ClinicDB, BKAI-IGH, and ETIS. Experimental findings demonstrate that the proposed method delivers higher segmentation accuracy in less time, consistently outperforming the most advanced lightweight polyp segmentation networks.

8.
Med Biol Eng Comput ; 62(7): 2087-2100, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38457066

RESUMEN

The pancreas not only is situated in a complex abdominal background but is also surrounded by other abdominal organs and adipose tissue, resulting in blurred organ boundaries. Accurate segmentation of pancreatic tissue is crucial for computer-aided diagnosis systems, as it can be used for surgical planning, navigation, and assessment of organs. In the light of this, the current paper proposes a novel Residual Double Asymmetric Convolution Network (ResDAC-Net) model. Firstly, newly designed ResDAC blocks are used to highlight pancreatic features. Secondly, the feature fusion between adjacent encoding layers fully utilizes the low-level and deep-level features extracted by the ResDAC blocks. Finally, parallel dilated convolutions are employed to increase the receptive field to capture multiscale spatial information. ResDAC-Net is highly compatible to the existing state-of-the-art models, according to three (out of four) evaluation metrics, including the two main ones used for segmentation performance evaluation (i.e., DSC and Jaccard index).


Asunto(s)
Páncreas , Humanos , Páncreas/diagnóstico por imagen , Procesamiento de Imagen Asistido por Computador/métodos , Algoritmos , Redes Neurales de la Computación
9.
Med Biol Eng Comput ; 62(6): 1673-1687, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38326677

RESUMEN

Early intervention in tumors can greatly improve human survival rates. With the development of deep learning technology, automatic image segmentation has taken a prominent role in the field of medical image analysis. Manually segmenting kidneys on CT images is a tedious task, and due to the diversity of these images and varying technical skills of professionals, segmentation results can be inconsistent. To address this problem, a novel ASD-Net network is proposed in this paper for kidney and kidney tumor segmentation tasks. First, the proposed network employs newly designed Adaptive Spatial-channel Convolution Optimization (ASCO) blocks to capture anisotropic information in the images. Then, other newly designed blocks, i.e., Dense Dilated Enhancement Convolution (DDEC) blocks, are utilized to enhance feature propagation and reuse it across the network, thereby improving its segmentation accuracy. To allow the network to segment complex and small kidney tumors more effectively, the Atrous Spatial Pyramid Pooling (ASPP) module is incorporated in its middle layer. With its generalized pyramid feature, this module enables the network to better capture and understand context information at various scales within the images. In addition to this, the concurrent spatial and channel squeeze & excitation (scSE) attention mechanism is adopted to better comprehend and manage context information in the images. Additional encoding layers are also added to the base (U-Net) and connected to the original encoding layer through skip connections. The resultant enhanced U-Net structure allows for better extraction and merging of high-level and low-level features, further boosting the network's ability to restore segmentation details. In addition, the combined Binary Cross Entropy (BCE)-Dice loss is utilized as the network's loss function. Experiments, conducted on the KiTS19 dataset, demonstrate that the proposed ASD-Net network outperforms the existing segmentation networks according to all evaluation metrics used, except for recall in the case of kidney tumor segmentation, where it takes the second place after Attention-UNet.


Asunto(s)
Procesamiento de Imagen Asistido por Computador , Neoplasias Renales , Riñón , Redes Neurales de la Computación , Humanos , Neoplasias Renales/diagnóstico por imagen , Neoplasias Renales/patología , Riñón/diagnóstico por imagen , Riñón/patología , Procesamiento de Imagen Asistido por Computador/métodos , Tomografía Computarizada por Rayos X/métodos , Aprendizaje Profundo , Algoritmos
10.
J Neurosci Res ; 91(2): 240-8, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23161705

RESUMEN

Damage to white matter such as corpus callosum (CC) is a pathological characteristic in many brain disorders. Glutamate (Glut) excitotoxicity through AMPA receptors on oligodendrocyte (OL) was previously considered as a mechanism for white matter damage. Recent studies have shown that N-methyl-D-aspartate receptors (NMDARs) are expressed on myelin sheath of neonatal rat OL processes and that activation of these receptors mediated demyelization. Whether NMDARs are expressed in the adult CC and are involved in excitotoxic axonal injury remains to be determined. In this study, we demonstrate the presence of NMDARs in the adult rat CC and their distributions in myelinated nerve fibers and OL somata by means of immunocytochemical staining and Western blot. Incubation of the CC slices with Glut or NMDA induced axonal injury as revealed by analyzing amplitude of CC fiber compound action potentials (CAPs) and input-output response. Both Glut and NMDA decreased the CAP amplitude and input-output responses, suggesting an involvement of NMDARs in Glut- and NMDA-induced axonal injury. The involvement of NMDAR in Glut-induced axonal injury was further assayed by detection of ß-amyloid precursor protein (ß-APP) in the CC axonal fibers. Treatment of the CC slices with Glut resulted in ß-APP accumulation in the CC fibers as detected by Western blot, reflecting an impairment of axonal transport function. This injurious effect of Glut on CC axonal transport was significantly blocked by MK801. Taken together, these results show that NMDARs are expressed in the adult CC and are involved in excitotoxic activity in adult CC slices in vitro.


Asunto(s)
Axones/patología , Cuerpo Calloso/metabolismo , Síndromes de Neurotoxicidad/patología , Receptores de N-Metil-D-Aspartato/metabolismo , Potenciales de Acción/efectos de los fármacos , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Modelos Animales de Enfermedad , Maleato de Dizocilpina/farmacología , Aminoácidos Excitadores/toxicidad , Femenino , Ácido Glutámico/toxicidad , Técnicas In Vitro , Masculino , Proteína Básica de Mielina/metabolismo , Fibras Nerviosas Mielínicas/patología , Fibras Nerviosas Mielínicas/fisiología , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Síndromes de Neurotoxicidad/etiología , Oligodendroglía/metabolismo , Técnicas de Placa-Clamp , Fosfopiruvato Hidratasa/metabolismo , Ratas , Ratas Sprague-Dawley
11.
Viruses ; 15(5)2023 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-37243203

RESUMEN

The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is the causative pathogen of the coronavirus disease 2019 (COVID-19) pandemic, a fatal respiratory illness. The associated risk factors for COVID-19 are old age and medical comorbidities. In the current combined antiretroviral therapy (cART) era, a significant portion of people living with HIV-1 (PLWH) with controlled viremia is older and with comorbidities, making these people vulnerable to SARS-CoV-2 infection and COVID-19-associated severe outcomes. Additionally, SARS-CoV-2 is neurotropic and causes neurological complications, resulting in a health burden and an adverse impact on PLWH and exacerbating HIV-1-associated neurocognitive disorder (HAND). The impact of SARS-CoV-2 infection and COVID-19 severity on neuroinflammation, the development of HAND and preexisting HAND is poorly explored. In the present review, we compiled the current knowledge of differences and similarities between SARS-CoV-2 and HIV-1, the conditions of the SARS-CoV-2/COVID-19 and HIV-1/AIDS syndemic and their impact on the central nervous system (CNS). Risk factors of COVID-19 on PLWH and neurological manifestations, inflammatory mechanisms leading to the neurological syndrome, the development of HAND, and its influence on preexisting HAND are also discussed. Finally, we have reviewed the challenges of the present syndemic on the world population, with a particular emphasis on PLWH.


Asunto(s)
COVID-19 , Infecciones por VIH , Seropositividad para VIH , VIH-1 , Enfermedades del Sistema Nervioso , Humanos , COVID-19/complicaciones , SARS-CoV-2 , Enfermedades del Sistema Nervioso/epidemiología , Enfermedades del Sistema Nervioso/etiología , Sistema Nervioso Central , Infecciones por VIH/complicaciones , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/epidemiología
12.
Artículo en Inglés | MEDLINE | ID: mdl-37457651

RESUMEN

Despite the introduction of combined antiretroviral therapy (cART) HIV-1 virus persists in the brain in a latent or restricted manner and viral proteins, such as gp120, continue to play a significant disease-inciting role. Gp120 is known to interact with N-methyl-D-aspartate (NMDA) receptors (NMDARs) resulting in neuronal injury. Glutamate is the main excitatory neurotransmitter in the brain and plays an important role in cognitive function and dysregulation of excitatory synaptic transmission impairs neurocognition. It is our hypothesis that gp120 may alter synaptic function via modulating glutamate function from a physiological molecule to a pathophysiological substance. To test this hypothesis, we studied the modulatory effects of gp120 and glutamate on NMDAR-mediated spontaneous excitatory postsynaptic current (sEPSCNMDAR) and dynamic dendritic spine changes in rat cortical neuronal cultures. Our results revealed that gp120 and glutamate each, at low concentrations, had no significant effects on sEPSCNMDAR and dendritic spines, but increased sEPSCNMDAR frequency, decreased numbers of dendritic spines when tested in combination. The observed effects were blocked by either a CXCR4 blocker or an NMDAR antagonist, indicating the involvements of chemokine receptor CXCR4 and NMDARs in gp120 modulation of glutamate effects. These results may imply a potential mechanism for HIV-1-associated neuropathogenesis in the cART era.

13.
Res Sq ; 2023 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-38168345

RESUMEN

Background: Human Immunodeficiency Virus type 1 (HIV-1)-associated neurocognitive disorders (HAND) remain prevalent in HIV-1-infected individuals despite the evident success of combined antiretroviral therapy (cART). The mechanisms under HAND prevalence in the cART era remain perplexing. Ample evidence indicates that HIV-1 envelope glycoprotein protein 120 (gp120), a potent neurotoxin, plays a pivotal role in the HAND pathogenesis. Methamphetamine (Meth) abuse exacerbates HAND. How Meth exacerbates HAND is not fully understood. This study was to test the hypothesis that Meth exacerbates HAND by enhancing gp120-mediated proinflammatory responses in the brain, worsening the pathogenesis of HAND. Methods: Experiments were carried out on primary microglial cultures prepared from neonatal SD rats. The purity of microglia was determined by staining with anti-CD11b. Meth and gp120 were applied to microglial cultures. Microglial activation was revealed by immunostaining and Iba-1 expression. The protein expression levels of Pro-IL-1ß, Il-1ß, Iba-1, iNOS, NLRP3, GSDMD and GSDMD-N were detected by western blotting analyses. The levels of proinflammatory cytokine and NO production in the microglia culture supernatants were assayed by ELISA and Griess reagent systems, respectively. NLRP3 activation was uncovered by fluorescent microscopy images displaying NLRP3 puncta labeled by anti-NLRP3 antibody. NLRP3 co-localization with caspase-1 was labeled with antibodies. One-way ANOVA with post hoc Tukey's multiple comparison tests was employed for statistical analyses. Results: Meth enhanced gp120-induced microglia activation revealed by immunostaining and Iba-1 expression, and potentiated gp120-mediated NLRP3 expression, IL-1ß processing and release assayed by immunoblot and ELISA. Meth also augmented the co-localization of NLRP3 and caspase-1, increased the numbers of NLRP3 puncta and ROS production, elevated levels of iNOS expression and NO production, and enhanced levels of cleaved gasderminD (GSDMD-N, an executor of pyroptosis) in gp120-primed microglia. The Meth-associated effects were attenuated or blocked by MCC950, an NLRP3 inhibitor, or Mito-TEMPO, a mitochondrial superoxide scavenger, indicating the involvement of mitochondria in Meth enhancement of NLRP3 inflammasome activation in gp120-primed microglia. Conclusions: These results suggest that Meth enhanced gp120-associated microglial NLRP3 activation and resultant proinflammatory responses via mitochondria-dependent signaling.

14.
Artículo en Inglés | MEDLINE | ID: mdl-35891930

RESUMEN

Despite the introduction of vaccines and drugs for SARS-CoV-2, the COVID-19 pandemic continues to spread throughout the world. In severe COVID-19 patients, elevated levels of proinflammatory cytokines have been detected in the blood, lung cells, and bronchoalveolar lavage, which is referred to as a cytokine storm, a consequence of overactivation of the NLR family pyrin domain-containing protein 3 (NLRP3) inflammasome and resultant excessive cytokine production. The hyperinflammatory response and cytokine storm cause multiorgan impairment including the central nervous system, in addition to a detriment to the respiratory system. Hyperactive NLRP3 inflammasome, due to dysregulated immune response, is the primary cause of COVID-19 severity. The severity could be enhanced due to viral evolution leading to the emergence of mutated variants of concern, such as delta and omicron. In this review, we elaborate on the inflammatory responses associated with the NLRP3 inflammasome activation in COVID-19 pathogenesis, the mechanisms for the NLRP3 inflammasome activation and pathway involved, cytokine storm, and neurological complications as long-term consequences of SARS-CoV-2 infection. Also discussed is the therapeutic potential of NLRP3 inflammasome inhibitors for the treatment of COVID-19.

15.
Glia ; 59(6): 997-1007, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21438014

RESUMEN

Microglia are critical cells in mediating the pathophysiology of neurodegenerative disorders such as HIV-associated neurocognitive disorders. We hypothesize that HIV-1 glycoprotein 120 (gp120) activates microglia by enhancing outward K(+) currents, resulting in microglia secretion of neurotoxins, consequent neuronal dysfunction, and death. To test this hypothesis, we studied the effects of gp120 on outward K(+) current in cultured rat microglia. Application of gp120 enhanced outward K(+) current in a dose-dependent manner, which was blocked by voltage-gated K(+) (K(v) ) channel blockers. Western blot analysis revealed that gp120 produced an elevated expression of K(v) channel proteins. Examination of activation and inactivation of outward K(+) currents showed that gp120 shifted membrane potentials for activation and steady-state inactivation. The gp120-associated enhancement of outward K(+) current was blocked by either a CXCR4 receptor antagonist T140 or a specific protein kinase A (PKA) inhibitor H89, suggesting the involvement of chemokine receptor CXCR4 and PKA in gp120-mediated enhancement of outward K(+) current. Biological significance of gp120-induced enhancement of microglia outward K(+) current was demonstrated by experimental results showing the neurotoxic activity of gp120-stimulated microglia, evaluated by TUNEL staining and MTT assay, significantly attenuated by K(v) channel blockers. Taken together, these results suggest that gp120 induces microglia neurotoxic activity by enhancing microglia outward K(+) current and that microglia K(v) channels may function as a potential target for the development of therapeutic strategies.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Proteína gp120 de Envoltorio del VIH/fisiología , Microglía/metabolismo , Microglía/virología , Canales de Potasio/metabolismo , Receptores CXCR4/fisiología , Transducción de Señal/fisiología , Complejo SIDA Demencia/enzimología , Complejo SIDA Demencia/metabolismo , Complejo SIDA Demencia/virología , Animales , Células Cultivadas , Técnicas de Cocultivo , Potenciales de la Membrana/fisiología , Microglía/enzimología , Degeneración Nerviosa/enzimología , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/virología , Ratas , Ratas Sprague-Dawley
16.
J Neurochem ; 116(3): 406-14, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21105875

RESUMEN

In addition to its well-characterized effects in immune system, chemokine CC motif ligand 2 (CCL2, formerly known as monocyte chemoattractant protein-1) is believed to play an important role in brain physiological and pathological processes. It has been shown that CCL2 and its cognate receptor chemokine CC motif receptor 2 are constitutively expressed in several brain regions including the hippocampus, and the expression is up-regulated under pathological conditions. Whereas most investigations have so far focused on its involvement in CNS pathology, few studies have examined the effects of CCL2 on neuronal and synaptic physiology. In this study, we tested the effects of CCL2 on neuronal excitability and excitatory synaptic transmission in the CA1 region of rat hippocampal slices using whole-cell patch clamp techniques. Bath application of CCL2 depolarized membrane potential and increased spike firing in CA1 neuronal cells. Bath application of CCL2 also produced an increase of excitatory post-synaptic currents recorded in Schaffer-collateral fibers to CA1 synapses. Quantal analysis revealed that CCL2 increased the frequency of spontaneous excitatory post-synaptic current occurrence and mean quantal content. Taken together, our data indicate that CCL2 enhances neuronal excitability and synaptic transmission via pre-synaptic mechanisms. These results support the emerging concept that chemokines function as neuromodulators in the CNS.


Asunto(s)
Potenciales de Acción/fisiología , Quimiocina CCL2/fisiología , Hipocampo/fisiología , Neuronas/fisiología , Transmisión Sináptica/fisiología , Animales , Axones/fisiología , Región CA1 Hipocampal/citología , Región CA1 Hipocampal/metabolismo , Potenciales Postsinápticos Excitadores/fisiología , Hipocampo/citología , Masculino , Neuronas/citología , Técnicas de Cultivo de Órganos , Terminales Presinápticos/fisiología , Ratas , Ratas Sprague-Dawley , Vesículas Sinápticas/fisiología
17.
J Exp Med ; 201(4): 567-77, 2005 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-15710649

RESUMEN

Decay-accelerating factor ([DAF] CD55) is a glycosylphosphatidylinositol-anchored membrane inhibitor of complement with broad clinical relevance. Here, we establish an additional and unexpected role for DAF in the suppression of adaptive immune responses in vivo. In both C57BL/6 and BALB/c mice, deficiency of the Daf1 gene, which encodes the murine homologue of human DAF, significantly enhanced T cell responses to active immunization. This phenotype was characterized by hypersecretion of interferon (IFN)-gamma and interleukin (IL)-2, as well as down-regulation of the inhibitory cytokine IL-10 during antigen restimulation of lymphocytes in vitro. Compared with wild-type mice, Daf1(-/-) mice also displayed markedly exacerbated disease progression and pathology in a T cell-dependent experimental autoimmune encephalomyelitis (EAE) model. However, disabling the complement system in Daf1(-/-) mice normalized T cell secretion of IFN-gamma and IL-2 and attenuated disease severity in the EAE model. These findings establish a critical link between complement and T cell immunity and have implications for the role of DAF and complement in organ transplantation, tumor evasion, and vaccine development.


Asunto(s)
Antígenos CD55/inmunología , Proteínas del Sistema Complemento/inmunología , Linfocitos T/inmunología , Animales , Antígenos CD55/genética , Antígenos CD55/metabolismo , Proliferación Celular , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Inmunización , Interferón gamma/biosíntesis , Interleucina-10/metabolismo , Interleucina-2/biosíntesis , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Bazo/citología , Bazo/inmunología
18.
J Immunol ; 182(6): 3855-65, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19265165

RESUMEN

HIV-1-associated neurocognitive impairments are intrinsically linked to microglial immune activation, persistent viral infection, and inflammation. In the era of antiretroviral therapy, more subtle cognitive impairments occur without adaptive immune compromise. We posit that adaptive immunity is neuroprotective, serving in both the elimination of infected cells through CD8(+) cytotoxic T cell activities and the regulation of neuroinflammatory responses of activated microglia. For the latter, little is known. Thus, we studied the neuromodulatory effects of CD4(+) regulatory T cells (Treg; CD4(+)CD25(+)) or effector T cells in HIV-1-associated neurodegeneration. A newly developed HIV-1 encephalitis mouse model was used wherein murine bone marrow-derived macrophages are infected with a full-length HIV-1(YU2)/vesicular stomatitis viral pseudotype and injected into basal ganglia of syngeneic immunocompetent mice. Adoptive transfer of CD3-activated Treg attenuated astrogliosis and microglia inflammation with concomitant neuroprotection. Moreover, Treg-mediated anti-inflammatory activities and neuroprotection were associated with up-regulation of brain-derived neurotrophic factor and glial cell-derived neurotrophic factor expression and down-regulation of proinflammatory cytokines, oxidative stress, and viral replication. Effector T cells showed contrary effects. These results, taken together, demonstrate the importance of Treg in disease control and raise the possibility of their utility for therapeutic strategies.


Asunto(s)
Complejo SIDA Demencia/prevención & control , Encefalitis Viral/prevención & control , Infecciones por VIH/prevención & control , VIH-1/inmunología , Subunidad alfa del Receptor de Interleucina-2/biosíntesis , Neuroinmunomodulación/inmunología , Linfocitos T Reguladores/inmunología , Complejo SIDA Demencia/inmunología , Complejo SIDA Demencia/patología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/patología , Células de la Médula Ósea/virología , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Encefalitis Viral/inmunología , Encefalitis Viral/patología , Infecciones por VIH/inmunología , Infecciones por VIH/patología , Macrófagos/inmunología , Macrófagos/patología , Macrófagos/virología , Ratones , Ratones Endogámicos C57BL , Microglía/inmunología , Microglía/patología , Microglía/virología , Factores de Crecimiento Nervioso/biosíntesis , Factores de Crecimiento Nervioso/fisiología , Linfocitos T Reguladores/patología , Linfocitos T Reguladores/virología , Estomatitis Vesicular/inmunología , Estomatitis Vesicular/patología , Estomatitis Vesicular/virología , Virus de la Estomatitis Vesicular Indiana/inmunología
19.
Eur J Neurosci ; 31(2): 214-22, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20074219

RESUMEN

Through their capacity to secrete, upon activation, a variety of bioactive molecules, brain macrophages (and resident microglia) play an important role in brain immune and inflammatory responses. To test our hypothesis that activated macrophages induce neuronal injury by enhancing neuronal outward K(+) current, we studied the effects of lipopolysaccharide (LPS)-stimulated human monocyte-derived macrophage (MDM) on neuronal transient A-type K(+) current (I(A)) and resultant neuronal injury in primary rat hippocampal neuronal cultures. Bath application of LPS-stimulated MDM-conditioned media (MCM+) enhanced neuronal I(A) in a concentration-dependent manner. Non-stimulated MCM (MCM-) failed to alter I(A). The enhancement of neuronal I(A) was recapitulated in neurons co-cultured with macrophages. The link of MCM(+)-induced enhancement of I(A) to MCM(+)-associated neuronal injury, as detected by propidium iodide and 4'',6-diamidino-2-phenylindol staining (DAPI) and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay, was demonstrated by experimental results showing that addition of I(A) blocker 4-aminopyridine to the cultures protected hippocampal neurons from MCM(+)-induced neuronal injury. Further investigation revealed that glutamate was involved in MCM(+)-induced enhancement of neuronal I(A). These results suggest that during brain inflammation macrophages (and microglia) might mediate neuronal injury via enhancement of neuronal I(A), and that neuronal K(v) channel might be a potential target for the development of therapeutic strategies for some neurodegenerative disorders by which immune and inflammatory responses are believed to be involved in the pathogenesis.


Asunto(s)
4-Aminopiridina/farmacología , Macrófagos/metabolismo , Neuronas/metabolismo , Neuronas/patología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/metabolismo , Animales , Células Cultivadas , Técnicas de Cocultivo , Medios de Cultivo Condicionados/metabolismo , Femenino , Humanos , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Monocitos/citología , Monocitos/metabolismo , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley
20.
Biochem Biophys Res Commun ; 402(2): 241-6, 2010 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-20933498

RESUMEN

Astrocytes play an important role in astrocyte-neuron homeostasis. In HIV-1-infected brain, interleukin 1 beta (IL-1ß) activation of astrocytes contributes to neurodegeneration. However, the molecular mechanisms underlying IL-1ß-activated-astrocytes-induced neurodegeneration in HIV-1-infected brain are largely unknown. We hypothesize that secretory factors from the activated astrocytes affect N-methyl-d-aspartate (NMDA) receptor, a major pathway implicated in HIV-1-associated neurodegeneration. To test this hypothesis, we studied effects of IL-1ß-stimulated astrocyte conditioned medium (ACM+) for its ability to activate NR1a/NR2B receptors expressed on Xenopus oocytes. Astrocytes treated with IL-1ß 20ng/ml for 24h induced CXCL8, CCL2, MMP1 and MMP7. Pressure ejection of the ACM(+) produced an inward current in NR1a/NR2B-expressing oocytes. The inward current produced by ACM(+) was blocked by NMDA receptor antagonist, APV but not by non-NMDA receptor antagonist, CNQX. These results suggest that IL-1ß stimulated astrocytes activate NR1a/NR2B receptors which may have implications in HIV-1-associated neurodegeneration.


Asunto(s)
Astrocitos/virología , VIH-1 , Enfermedades Neurodegenerativas/virología , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Astrocitos/metabolismo , Células Cultivadas , Quimiocina CCL2/metabolismo , Quimiocina CCL2/farmacología , Medios de Cultivo Condicionados/metabolismo , Medios de Cultivo Condicionados/farmacología , Humanos , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacología , Interleucina-8/metabolismo , Interleucina-8/farmacología , Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 1 de la Matriz/farmacología , Metaloproteinasa 7 de la Matriz/metabolismo , Metaloproteinasa 7 de la Matriz/farmacología , Enfermedades Neurodegenerativas/metabolismo , Oocitos , Xenopus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA