Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(12)2021 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-34203972

RESUMEN

Opioid abuse has become a major public health crisis that affects millions of individuals across the globe. This widespread abuse of prescription opioids and dramatic increase in the availability of illicit opioids have created what is known as the opioid epidemic. Pregnant women are a particularly vulnerable group since they are prescribed for opioids such as morphine, buprenorphine, and methadone, all of which have been shown to cross the placenta and potentially impact the developing fetus. Limited information exists regarding the effect of oxycodone (oxy) on synaptic alterations. To fill this knowledge gap, we employed an integrated system approach to identify proteomic signatures and pathways impacted on mixed neuroglial cultures treated with oxy for 24 h. Differentially expressed proteins were mapped onto global canonical pathways using ingenuity pathway analysis (IPA), identifying enriched pathways associated with ephrin signaling, semaphorin signaling, synaptic long-term depression, endocannabinoid signaling, and opioid signaling. Further analysis by ClueGO identified that the dominant category of differentially expressed protein functions was associated with GDP binding. Since opioid receptors are G-protein coupled receptors (GPCRs), these data indicate that oxy exposure perturbs key pathways associated with synaptic function.


Asunto(s)
Neuroglía/metabolismo , Oxicodona/farmacología , Proteoma/metabolismo , Análisis de Sistemas , Animales , Muerte Celular/efectos de los fármacos , Células Cultivadas , Ontología de Genes , Neuroglía/efectos de los fármacos , Proteómica , Ratas Sprague-Dawley
3.
Int J Mol Sci ; 21(18)2020 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-32942668

RESUMEN

Extracellular vesicles (EVs) are a broad, heterogeneous class of membranous lipid-bilayer vesicles that facilitate intercellular communication throughout the body. As important carriers of various types of cargo, including proteins, lipids, DNA fragments, and a variety of small noncoding RNAs, including miRNAs, mRNAs, and siRNAs, EVs may play an important role in the development of addiction and other neurological pathologies, particularly those related to HIV. In this review, we summarize the findings of EV studies in the context of methamphetamine (METH), cocaine, nicotine, opioid, and alcohol use disorders, highlighting important EV cargoes that may contribute to addiction. Additionally, as HIV and substance abuse are often comorbid, we discuss the potential role of EVs in the intersection of substance abuse and HIV. Taken together, the studies presented in this comprehensive review shed light on the potential role of EVs in the exacerbation of substance use and HIV. As a subject of growing interest, EVs may continue to provide information about mechanisms and pathogenesis in substance use disorders and CNS pathologies, perhaps allowing for exploration into potential therapeutic options.


Asunto(s)
Vesículas Extracelulares/metabolismo , Infecciones por VIH/metabolismo , Enfermedades del Sistema Nervioso/metabolismo , Trastornos Relacionados con Sustancias/metabolismo , Animales , Humanos
4.
Can J Neurol Sci ; 45(2): 176-181, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29307324

RESUMEN

BACKGROUND: Recent advances in neurophysiological techniques have contributed to our understanding of the pathophysiology of Huntington's disease (HD). Studies of the motor cortical excitability and central motor pathways have shown variable results. OBJECTIVES: Our aims were to evaluate the cortical excitability changes in HD using transcranial magnetic stimulation (TMS) and correlate the changes with cognitive impairment. METHODS: The study included 32 HD patients and 30 age- and gender-matched controls. The demographic and clinical profiles of the patients were recorded. All subjects were evaluated by TMS and resting motor threshold (RMT), central motor conduction time (CMCT), silent period (SP), short-interval intracortical inhibition (SICI), and intracortical facilitation were determined. A battery of neuropsychological tests was administered to all subjects. RESULTS: The mean age of the patients was 42.1±14.1 years, and that of controls 39.4±12.4 years (p=0.61). There was no significant difference in RMT and CMCT between the two groups. There was a mild prolongation of the contralateral SP in HD, but it was not significant. SICI was significantly reduced in HD (p<0.0001). A significant impairment in attention, verbal fluency, executive function, visuospatial function, learning, and memory was observed in HD patients. However, there was no correlation between cortical excitability changes and cognitive impairment. CONCLUSIONS: TMS is a valuable method of evaluating cortical excitability changes in HD. These patients have reduced SICI and significant impairment of cognition in multiple domains.


Asunto(s)
Corteza Cerebral/fisiopatología , Trastornos del Conocimiento/etiología , Enfermedad de Huntington/complicaciones , Enfermedad de Huntington/patología , Adulto , Estudios de Casos y Controles , Correlación de Datos , Electromiografía , Función Ejecutiva , Femenino , Humanos , Enfermedad de Huntington/terapia , Masculino , Persona de Mediana Edad , Inhibición Neural/fisiología , Pruebas Neuropsicológicas , Estimulación Magnética Transcraneal , Resultado del Tratamiento , Aprendizaje Verbal , Percepción Visual
5.
PLoS Pathog ; 11(7): e1005032, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26154133

RESUMEN

Recent studies have found that extracellular vesicles (EVs) play an important role in normal and disease processes. In the present study, we isolated and characterized EVs from the brains of rhesus macaques, both with and without simian immunodeficiency virus (SIV) induced central nervous system (CNS) disease. Small RNA sequencing revealed increased miR-21 levels in EVs from SIV encephalitic (SIVE) brains. In situ hybridization revealed increased miR-21 expression in neurons and macrophage/microglial cells/nodules during SIV induced CNS disease. In vitro culture of macrophages revealed that miR-21 is released into EVs and is neurotoxic when compared to EVs derived from miR-21-/- knockout animals. A mutation of the sequence within miR-21, predicted to bind TLR7, eliminates this neurotoxicity. Indeed miR-21 in EV activates TLR7 in a reporter cell line, and the neurotoxicity is dependent upon TLR7, as neurons isolated from TLR7-/- knockout mice are protected from neurotoxicity. Further, we show that EVs isolated from the brains of monkeys with SIV induced CNS disease activates TLR7 and were neurotoxic when compared to EVs from control animals. Finally, we show that EV-miR-21 induced neurotoxicity was unaffected by apoptosis inhibition but could be prevented by a necroptosis inhibitor, necrostatin-1, highlighting the actions of this pathway in a growing number of CNS disorders.


Asunto(s)
Vesículas Extracelulares/metabolismo , MicroARNs/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/genética , Receptor Toll-Like 7/metabolismo , Animales , Western Blotting , Encéfalo/virología , Técnica del Anticuerpo Fluorescente , Hibridación Fluorescente in Situ , Macaca mulatta , Macrófagos/metabolismo , Macrófagos/virología , Ratones , Ratones Noqueados , Microscopía Electrónica de Transmisión , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Virus de la Inmunodeficiencia de los Simios/genética
6.
FASEB J ; 27(9): 3720-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23752207

RESUMEN

MicroRNA (miR)-142 is up-regulated in the brain in HIV and SIV encephalitis (SIVE). We identified the cell types where miR-142 is up-regulated and its relevant downstream target. Fluorescent in situ hybridization combined with immunofluorescent labeling revealed that miR-142-3p and -5p are expressed within hippocampal neurons and myeloid cells in SIVE. Sirtuin1 (SIRT1) was predicted as a potential miR-142 target by analysis of its 3'-UTR and bioinformatic analysis of factors linked to altered hippocampal gene expression profile in SIVE. Overexpression of pre-miR-142 in HEK293T cells led to a 3.7-fold decrease in SIRT1 protein level. Examination of the individual effects of miR-142-5p and miR-142-3p through overexpression and inhibition studies revealed that significant effects on SIRT1 occurred only with miR-142-5p. Luciferase reporter assays revealed a 2.3-fold inhibition of expression due to interaction of miR-142 with the SIRT1 3'-UTR, mutation analysis revealed that only the miR-142-5p target site was active. MiR-142 expression in primary human neurons led to a small (1.3-fold) but significant decrease in SIRT1 protein level. Furthermore, qRT-PCR revealed up-regulation of miR-142-3p (6.4-fold) and -5p (3.9-fold) and down-regulation of SIRT1 (33-fold) in macrophages/microglia from animals with SIVE. We have therefore elucidated a miR-mediated mechanism of regulation of SIRT1 expression in SIVE.


Asunto(s)
Encefalitis/genética , Encefalitis/virología , MicroARNs/genética , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Sirtuina 1/metabolismo , Animales , Western Blotting , Línea Celular , Células Cultivadas , Humanos , Sirtuina 1/genética , Regulación hacia Arriba
7.
J Neuroimmune Pharmacol ; 19(1): 29, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38874861

RESUMEN

The opioid epidemic has received considerable attention, but the impact on perinatal opioid-exposed (POE) offspring remains underexplored. This study addresses the emerging public health challenge of understanding and treating POE children. We examined two scenarios using preclinical models: offspring exposed to oxycodone (OXY) in utero (IUO) and acute postnatal OXY (PNO). We hypothesized exposure to OXY during pregnancy primes offspring for neurodevelopmental deficits and severity of deficits is dependent on timing of exposure. Notable findings include reduced head size and brain weight in offspring. Molecular analyses revealed significantly lower levels of inflammasome-specific genes in the prefrontal cortex (PFC). Gene Set Enrichment Analysis (GSEA) and Ingenuity Pathway Analysis (IPA) highlighted the enrichment of genes associated with mitochondrial and synapse dysfunction in POE offspring. Western blot analysis validated IPA predictions of mitochondrial dysfunction in PFC-derived synaptosomes. Behavioral studies identified significant social deficits in POE offspring. This study presents the first comparative analysis of acute PNO- and IUO-offspring during early adolescence finding acute PNO-offspring have considerably greater deficits. The striking difference in deficit severity in acute PNO-offspring suggests that exposure to opioids in late pregnancy pose the greatest risk for offspring well-being.


Asunto(s)
Analgésicos Opioides , Oxicodona , Efectos Tardíos de la Exposición Prenatal , Animales , Oxicodona/toxicidad , Embarazo , Femenino , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Masculino , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/toxicidad , Conducta Animal/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Trastornos del Neurodesarrollo/inducido químicamente , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo
8.
ACS Omega ; 9(3): 3164-3172, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38284070

RESUMEN

Over the past few decades, it has been well established that gut microbiota-derived metabolites can disrupt gut function, thus resulting in an array of diseases. Notably, phenylacetylglutamine (PAGln), a bacterial derived metabolite, has recently gained attention due to its role in the initiation and progression of cardiovascular and cerebrovascular diseases. This meta-organismal metabolite PAGln is a byproduct of amino acid acetylation of its precursor phenylacetic acid (PAA) from a range of dietary sources like egg, meat, dairy products, etc. The microbiota-dependent metabolism of phenylalanine produces PAA, which is a crucial intermediate that is catalyzed by diverse microbial catalytic pathways. PAA conjugates with glutamine and glycine in the liver and kidney to predominantly form phenylacetylglutamine in humans and phenylacetylglycine in rodents. PAGln is associated with thrombosis as it enhances platelet activation mediated through the GPCRs receptors α2A, α2B, and ß2 ADRs, thereby aggravating the pathological conditions. Clinical evidence suggests that elevated levels of PAGln are associated with pathology of cardiovascular, cerebrovascular, and neurological diseases. This Review further consolidates the microbial/biochemical synthesis of PAGln and discusses its role in the above pathophysiologies.

9.
Orbit ; 32(6): 392-4, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24063411

RESUMEN

Abstract Esthesioneuroblastoma, also known as olfactory neuroblastoma, is an uncommon malignant neoplasm arising from the olfactory epithelium in the roof of the nasal cavity. There are very few case reports published worldwide. The common presenting symptoms of Esthesioneuroblastoma are unilateral nasal obstruction (70%), epistaxis (50%), anosmia, rhinorrhoea, facial pain, headache, excessive lacrimation and rarely proptosis and visual disturbance. Apart from being locally aggressive, it metastasizes by haematogenous and lymphatic routes. We report an extremely rare case of esthesioneuroblastoma in a 20-year-old man with orbital involvement presenting as dystopia. This rare tumour should be considered in the differential diagnosis for young patients presenting to ophthalmic outpatient department with dystopia.


Asunto(s)
Estesioneuroblastoma Olfatorio/diagnóstico , Anomalías del Ojo/diagnóstico , Cavidad Nasal/patología , Neoplasias Nasales/diagnóstico , Órbita/anomalías , Terapia Combinada , Craneotomía , Diagnóstico Diferencial , Estesioneuroblastoma Olfatorio/radioterapia , Estesioneuroblastoma Olfatorio/cirugía , Exoftalmia , Humanos , Imagen por Resonancia Magnética , Masculino , Neoplasias Nasales/radioterapia , Neoplasias Nasales/cirugía , Tomografía Computarizada por Rayos X , Adulto Joven
10.
Neural Comput Appl ; 35(11): 8259-8279, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36532883

RESUMEN

Pneumonia is an acute respiratory infection caused by bacteria, viruses, or fungi and has become very common in children ranging from 1 to 5 years of age. Common symptoms of pneumonia include difficulty breathing due to inflamed or pus and fluid-filled alveoli. The United Nations Children's Fund reports nearly 800,000 deaths in children due to pneumonia. Delayed diagnosis and overpriced tests are the prime reason for the high mortality rate, especially in underdeveloped countries. A time and cost-efficient diagnosis tool: Chest X-rays, was thus accepted as the standard diagnostic test for pediatric pneumonia. However, the lower radiation levels for diagnosis in children make the task much more onerous and time-consuming. The mentioned challenges initiate the need for a computer-aided detection model that is instantaneous and accurate. Our work proposes a stacked ensemble learning of deep learning-based features for pediatric pneumonia classification. The extracted features from the global average pooling layer of the fine-tuned Xception model pretrained on ImageNet weights are sent to the Kernel Principal Component Analysis for dimensionality reduction. The dimensionally reduced features are further trained and validated on the stacking classifier. The stacking classifier consists of two stages; the first stage uses the Random-Forest classifier, K-Nearest Neighbors, Logistic Regression, XGB classifier, Support Vector Classifier (SVC), Nu-SVC, and MLP classifier. The second stage operates on Logistic Regression using the first stage predictions for the final classification with Stratified K-fold cross-validation to prevent overfitting. The model was tested on the publicly available pediatric pneumonia dataset, achieving an accuracy of 98.3%, precision of 99.29%, recall of 98.36%, F1-score of 98.83%, and an AUC score of 98.24%. The performance shows its reliability for real-time deployment in assisting radiologists and physicians.

11.
Multimed Tools Appl ; 82(14): 21311-21351, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36281318

RESUMEN

Pediatric pneumonia has drawn immense awareness due to the high mortality rates over recent years. The acute respiratory infection caused by bacteria, viruses, or fungi infects the lung region and hinders oxygen transport, making breathing difficult due to inflamed or pus and fluid-filled alveoli. Being non-invasive and painless, chest X-rays are the most common modality for pediatric pneumonia diagnosis. However, the low radiation levels for diagnosis in children make accurate detection challenging. This challenge initiates the need for an unerring computer-aided diagnosis model. Our work proposes Contrast Limited Adaptive Histogram Equalization for image enhancement and a stacking classifier based on the fusion of deep learning-based features for pediatric pneumonia diagnosis. The extracted features from the global average pooling layers of the fine-tuned MobileNet, DenseNet121, DenseNet169, and DenseNet201 are concatenated for the final classification using a stacked ensemble classifier. The stacking classifier uses Support Vector Classifier, Nu-SVC, Logistic Regression, K-Nearest Neighbor, Random Forest Classifier, Gaussian Naïve Bayes, AdaBoost classifier, Bagging Classifier, and Extra-trees Classifier for the first stage, and Nu-SVC as the meta-classifier. The stacking classifier validated using Stratified K-Fold cross-validation achieves an accuracy of 98.62%, precision of 98.99%, recall of 99.53%, F1 score of 99.26%, and an AUC score of 93.17% on the publicly available pediatric pneumonia dataset. We expect this model to greatly help the real-time diagnosis of pediatric pneumonia.

12.
Brain Behav Immun Health ; 32: 100669, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37588011

RESUMEN

Maternal opioid use poses a significant health concern not just to the expectant mother but also to the fetus. Notably, increasing numbers of children born suffering from neonatal opioid withdrawal syndrome (NOWS) further compounds the crisis. While epidemiological research has shown the heightened risk factors associated with NOWS, little research has investigated what molecular mechanisms underly the vulnerabilities these children carry throughout development and into later life. To understand the implications of in utero and post-natal opioid exposure on the developing brain, we sought to assess the response to one of the most common pediatric injuries: minor traumatic brain injury (mTBI). Using a rat model of in utero and post-natal oxycodone (IUO) exposure and a low force weight drop model of mTBI, we show that not only neonatal opioid exposure significantly affects neuroinflammation, brain metabolites, synaptic proteome, mitochondrial function, and altered behavior in juvenile rats, but also, in conjunction with mTBI these aberrations are further exacerbated. Specifically, we observed long term metabolic dysregulation, neuroinflammation, alterations in synaptic mitochondria, and impaired behavior were impacted severely by mTBI. Our research highlights the specific vulnerability caused by IUO exposure to a secondary stressor such as later life brain injury. In summary, we present a comprehensive study to highlight the damaging effects of prenatal opioid abuse in conjunction with mild brain injury on the developing brain.

13.
Res Sq ; 2023 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-37066266

RESUMEN

Perinatal exposure to prescription opioids pose a critical public health risk. Notably, research has found significant neurodevelopmental and behavioral deficits between in utero (IUO) and postnatal (PNO) oxycodone-exposed offspring but there is a notable gap in knowledge regarding the interaction of these groups to other drug exposure, particularly nicotine exposure. Nicotine's widespread use represents a ubiquitous clinical interaction that current research does not address. Children often experiment with drugs and risky behavior; therefore, adolescence is a key timepoint to characterize. This study employed an integrated systems approach to investigate escalating nicotine exposure in adolescence and subsequent nicotine withdrawal in the IUO- and PNO-offspring. Western blot analysis found alterations of the blood-brain barrier (B.B.B.) and synaptic proteins. RT-qPCR further validated immune dysfunction in the central nervous system (CNS) consistent with compromised B.B.B. Peripheral nicotine metabolism was consistent with increased catabolism of nicotine concerning PNO & IUO, a predictor of greater addiction risk. Lastly, behavioral assays found subtle deficits to withdrawal in nociception and anxiety-like behavior. This study showed, for the first time, the vulnerabilities of PNO- and IUO-exposed groups concerning nicotine use during early adolescence and withdrawal.

14.
J Neuroimmune Pharmacol ; 18(3): 413-426, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37351737

RESUMEN

Perinatal exposure to prescription opioids pose a critical public health risk. Notably, research has found significant neurodevelopmental and behavioral deficits between in utero (IUO) and postnatal (PNO) oxycodone-exposed offspring but there is a notable gap in knowledge regarding the interaction of these groups to other drug exposure, particularly nicotine exposure. Nicotine's widespread use represents a ubiquitous clinical interaction that current research does not address. Children often experiment with drugs and risky behavior; therefore, adolescence is a key timepoint to characterize. This study employed an integrated systems approach to investigate escalating nicotine exposure in adolescence and subsequent nicotine withdrawal in the IUO- and PNO-offspring. Western blot analysis found synaptic protein alterations, especially upregulation of synaptophysin in IUO-withdrawal animals. RT-qPCR further validated immune dysfunction in the central nervous system (CNS). Peripheral nicotine metabolism was consistent with increased catabolism of nicotine concerning IUO animals. Lastly, behavioral assays found subtle deficits to withdrawal in nociception and anxiety-like behavior. This study showed, for the first time, the vulnerabilities of PNO- and IUO-exposed groups concerning nicotine use during early adolescence and withdrawal. Graphical Abstract.


Asunto(s)
Nicotina , Agonistas Nicotínicos , Embarazo , Animales , Femenino , Niño , Humanos , Adolescente , Nicotina/toxicidad , Agonistas Nicotínicos/toxicidad , Oxicodona/efectos adversos
15.
Cells ; 12(6)2023 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-36980307

RESUMEN

Recently, the long-term use of sedative agents in the neonatal intensive care unit (NICU) has raised concerns about neurodevelopmental outcomes in exposed neonates. Midazolam (MDZ), a common neonatal sedative in the NICU, has been suggested to increase learning disturbances and cognitive impairment in children. However, molecular mechanisms contributing to such outcomes with long-term MDZ use during the early stages of life remain unclear. In this study, we for the first time elucidate the role of brain-derived extracellular vesicles (BDEVs), including mining the BDEV proteome post long-term MDZ exposure during early development. Employing our previously established rodent model system that mimics the exposure of MDZ in the NICU using an increasing dosage regimen, we isolated BDEVs from postnatal 21-days-old control and MDZ groups using a differential sucrose density gradient. BDEVs from the control and MDZ groups were then characterized using a ZetaView nanoparticle tracking analyzer and transmission electron microscopy analysis. Next, using RT-qPCR, we examined the expression of key ESCRT-related genes involved in EV biogenesis. Lastly, using quantitative mass spectrometry-based proteomics, we mined the BDEV protein cargo that revealed key differentially expressed proteins and associated molecular pathways to be altered post long-term MDZ exposure. Our study characterized the proteome in BDEV cargo from long-term MDZ exposure at early development. Importantly, we identified and validated the expression of YWHAH as a potential target for further characterization of its downstream mechanism and a potential biomarker for the early onset of neurodevelopment and neurodegenerative diseases. Overall, the present study demonstrated long-term exposure to MDZ at early development stages could influence BDEV protein cargo, which potentially impact neural functions and behavior at later stages of development.


Asunto(s)
Proteínas 14-3-3 , Vesículas Extracelulares , Midazolam , Animales , Ratas , Biomarcadores , Encéfalo , Vesículas Extracelulares/metabolismo , Hipnóticos y Sedantes/efectos adversos , Midazolam/efectos adversos , Midazolam/farmacología , Modelos Biológicos , Proteoma
16.
Viruses ; 15(9)2023 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-37766354

RESUMEN

In the 21st century, the effects of HIV-associated neurocognitive disorders (HAND) have been significantly reduced in individuals due to the development of antiretroviral therapies (ARTs). However, the growing epidemic of polysubstance use (PSU) has led to concern for the effects of PSU on HIV-seropositive individuals. To effectively treat individuals affected by HAND, it is critical to understand the biological mechanisms affected by PSU, including the identification of novel markers. To fill this important knowledge gap, we used an in vivo HIV-1 Transgenic (HIV-1 Tg) animal model to investigate the effects of the combined use of chronic methamphetamine (METH) and oxycodone (oxy). A RNA-Seq analysis on the striatum-a brain region that is primarily targeted by both HIV and drugs of abuse-identified key differentially expressed markers post-METH and oxy exposure. Furthermore, ClueGO analysis and Ingenuity Pathway Analysis (IPA) revealed crucial molecular and biological functions associated with ATP-activated adenosine receptors, neuropeptide hormone activity, and the oxytocin signaling pathway to be altered between the different treatment groups. The current study further reveals the harmful effects of chronic PSU and HIV infection that can subsequently impact neurological outcomes in polysubstance users with HAND.


Asunto(s)
Infecciones por VIH , VIH-1 , Metanfetamina , Animales , Humanos , Infecciones por VIH/complicaciones , Infecciones por VIH/tratamiento farmacológico , Oxicodona/farmacología , RNA-Seq , Trastornos Neurocognitivos , VIH-1/genética , Metanfetamina/farmacología
18.
Viruses ; 14(3)2022 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-35336957

RESUMEN

Despite the success of combinational antiretroviral therapy (cART), the high pervasiveness of human immunodeficiency virus-1 (HIV)-associated neurocognitive disorders (HAND) poses a significant challenge for society. Methamphetamine (meth) and related amphetamine compounds, which are potent psychostimulants, are among the most commonly used illicit drugs. Intriguingly, HIV-infected individuals who are meth users have a comparatively higher rate of neuropsychological impairment and exhibit a higher viral load in the brain than infected individuals who do not abuse meth. Effectively, all cell types secrete nano-sized lipid membrane vesicles, referred to as extracellular vesicles (EVs) that can function as intercellular communication to modulate the physiology and pathology of the cells. This study shows that meth treatments on chronically HIV-infected promonocytic U1 cells induce the release of EVs that promote cellular clustering and syncytia formation, a phenomenon that facilitates HIV pathogenesis. Our analysis also revealed that meth exposure increased intercellular adhesion molecule-1 (ICAM-1) and HIV-Nef protein expression in both large (10 K) and small (100 K) EVs. Further, when meth EVs are applied to uninfected naïve monocyte-derived macrophages (MDMs), we saw a significant increase in cell clustering and syncytia formation. Furthermore, treatment of MDMs with antibodies against ICAM-1 and its receptor, lymphocyte function-associated antigen 1 (LFA1), substantially blocked syncytia formation, and consequently reduced the number of multinucleated cells. In summary, our findings reveal that meth exacerbates HIV pathogenesis in the brain through release of proadhesive EVs, promoting syncytia formation and thereby aiding in the progression of HIV infection in uninfected cells.


Asunto(s)
Vesículas Extracelulares , Infecciones por VIH , VIH-1 , Metanfetamina , Vesículas Extracelulares/metabolismo , Células Gigantes , VIH-1/fisiología , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Metanfetamina/farmacología
19.
Genes (Basel) ; 13(10)2022 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-36292701

RESUMEN

Polysubstance use (PSU) generally involves the simultaneous use of an opioid along with a stimulant. In recent years, this problem has escalated into a nationwide epidemic. Understanding the mechanisms and effects underlying the interaction between these drugs is essential for the development of treatments for those suffering from addiction. Currently, the effect of PSU on synapses-critical points of contact between neurons-remains poorly understood. Using an in vitro model of primary neurons, we examined the combined effects of the psychostimulant methamphetamine (METH) and the prescription opioid oxycodone (oxy) on the synaptic proteome using quantitative mass-spectrometry-based proteomics. A further ClueGO analysis and Ingenuity Pathway Analysis (IPA) indicated the dysregulation of several molecular functions, biological processes, and pathways associated with neural plasticity and structural development. We identified one key synaptic protein, Striatin-1, which plays a vital role in many of these processes and functions, to be downregulated following METH+oxy treatment. This downregulation of Striatin-1 was further validated by Western blot. Overall, the present study indicates several damaging effects of the combined use of METH and oxy on neural function and warrants further detailed investigation into mechanisms contributing to synaptic dysfunction.


Asunto(s)
Estimulantes del Sistema Nervioso Central , Metanfetamina , Metanfetamina/farmacología , Oxicodona/farmacología , Proteoma/genética , Analgésicos Opioides , Estimulantes del Sistema Nervioso Central/farmacología
20.
Cells ; 11(11)2022 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-35681434

RESUMEN

The current opioid crisis, which has ravaged all segments of society, continues to pose a rising public health concern. Importantly, dependency on prescription opioids such as oxycodone (oxy) during and after pregnancy can significantly impact the overall brain development of the exposed offspring, especially at the synapse. A significant knowledge gap that remains is identifying distinct synaptic signatures associated with these exposed offspring. Accordingly, the overall goal of this current study was to identify distinct synaptic vesicle (SV) proteins as signatures for offspring exposed to oxy in utero (IUO) and postnatally (PNO). Using a preclinical animal model that imitates oxycodone exposure in utero (IUO) and postnatally (PNO), we used a quantitative mass spectrometry-based proteomics platform to examine changes in the synaptic vesicle proteome on post-natal day 14 (P14) IUO and PNO offspring. We identified MEGF8, associated with carpenter syndrome, to be downregulated in the IUO offspring while LAMTOR4, associated with the regulator complex involved in lysosomal signaling and trafficking, was found to be upregulated in the PNO groups, respectively. Their respective differential expression was further validated by Western blot. In summary, our current study shows exposure to oxy in utero and postnatally can impact the SV proteome in the exposed offspring and the identification of these distinct SV signatures could further pave the way to further elucidate their downstream mechanisms including developing them as potential therapeutic targets.


Asunto(s)
Oxicodona , Proteómica , Vesículas Sinápticas , Animales , Femenino , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Oxicodona/farmacología , Embarazo , Efectos Tardíos de la Exposición Prenatal , Proteoma/metabolismo , Sinapsis/metabolismo , Vesículas Sinápticas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA