Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(28): e2219543120, 2023 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-37406092

RESUMEN

Although HIV-1 Gag is known to drive viral assembly and budding, the precise mechanisms by which the lipid composition of the plasma membrane is remodeled during assembly are incompletely understood. Here, we provide evidence that the sphingomyelin hydrolase neutral sphingomyelinase 2 (nSMase2) interacts with HIV-1 Gag and through the hydrolysis of sphingomyelin creates ceramide that is necessary for proper formation of the viral envelope and viral maturation. Inhibition or depletion of nSMase2 resulted in the production of noninfectious HIV-1 virions with incomplete Gag lattices lacking condensed conical cores. Inhibition of nSMase2 in HIV-1-infected humanized mouse models with a potent and selective inhibitor of nSMase2 termed PDDC [phenyl(R)-(1-(3-(3,4-dimethoxyphenyl)-2, 6-dimethylimidazo[1,2-b]pyridazin-8-yl) pyrrolidin-3-yl)-carbamate] produced a linear reduction in levels of HIV-1 in plasma. If undetectable plasma levels of HIV-1 were achieved with PDDC treatment, viral rebound did not occur for up to 4 wk when PDDC was discontinued. In vivo and tissue culture results suggest that PDDC selectively kills cells with actively replicating HIV-1. Collectively, this work demonstrates that nSMase2 is a critical regulator of HIV-1 replication and suggests that nSMase2 could be an important therapeutic target with the potential to kill HIV-1-infected cells.


Asunto(s)
VIH-1 , Esfingomielina Fosfodiesterasa , Ratones , Animales , Esfingomielina Fosfodiesterasa/metabolismo , VIH-1/metabolismo , Esfingomielinas/metabolismo , Membrana Celular/metabolismo
2.
Proc Natl Acad Sci U S A ; 120(28): e2219475120, 2023 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-37406093

RESUMEN

HIV-1 assembly occurs at the inner leaflet of the plasma membrane (PM) in highly ordered membrane microdomains. The size and stability of membrane microdomains is regulated by activity of the sphingomyelin hydrolase neutral sphingomyelinase 2 (nSMase2) that is localized primarily to the inner leaflet of the PM. In this study, we demonstrate that pharmacological inhibition or depletion of nSMase2 in HIV-1-producer cells results in a block in the processing of the major viral structural polyprotein Gag and the production of morphologically aberrant, immature HIV-1 particles with severely impaired infectivity. We find that disruption of nSMase2 also severely inhibits the maturation and infectivity of other primate lentiviruses HIV-2 and simian immunodeficiency virus, has a modest or no effect on nonprimate lentiviruses equine infectious anemia virus and feline immunodeficiency virus, and has no effect on the gammaretrovirus murine leukemia virus. These studies demonstrate a key role for nSMase2 in HIV-1 particle morphogenesis and maturation.


Asunto(s)
VIH-1 , Virus de la Anemia Infecciosa Equina , Animales , Gatos , Caballos , Ratones , VIH-1/fisiología , Esfingomielina Fosfodiesterasa/metabolismo , Ensamble de Virus , Lentivirus
3.
Neurobiol Dis ; 177: 105987, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36603748

RESUMEN

Extracellular vesicles (EVs) have been proposed to regulate the deposition of Aß. Multiple publications have shown that APP, amyloid processing enzymes and Aß peptides are associated with EVs. However, very little Aß is associated with EVs compared with the total amount Aß present in human plasma, CSF, or supernatants from cultured neurons. The involvement of EVs has largely been inferred by pharmacological inhibition or whole body deletion of the sphingomyelin hydrolase neutral sphingomyelinase-2 (nSMase2) that is a key regulator for the biogenesis of at-least one population of EVs. Here we used a Cre-Lox system to selectively delete nSMase2 from pyramidal neurons in APP/PS1 mice (APP/PS1-SMPD3-Nex1) and found a âˆ¼ 70% reduction in Aß deposition at 6 months of age and âˆ¼ 35% reduction at 12 months of age in both cortex and hippocampus. Brain ceramides were increased in APP/PS1 compared with Wt mice, but were similar to Wt in APP/PS1-SMPD3-Nex1 mice suggesting that elevated brain ceramides in this model involves neuronally expressed nSMase2. Reduced levels of PSD95 and deficits of long-term potentiation in APP/PS1 mice were normalized in APP/PS1-SMPD3-Nex1 mice. In contrast, elevated levels of IL-1ß, IL-8 and TNFα in APP/PS1 mice were not normalized in APP/PS1-SMPD3-Nex1 mice compared with APP/PS1 mice. Mechanistic studies showed that the size of liquid ordered membrane microdomains was increased in APP/PS1 mice, as were the amounts of APP and BACE1 localized to these microdomains. Pharmacological inhibition of nSMase2 activity with PDDC reduced the size of the liquid ordered membrane microdomains, reduced the localization of APP with BACE1 and reduced the production of Aß1-40 and Aß1-42. Although inhibition of nSMase2 reduced the release and increased the size of EVs, very little Aß was associated with EVs in all conditions tested. We also found that nSMase2 directly protected neurons from the toxic effects of oligomerized Aß and preserved neural network connectivity despite considerable Aß deposition. These data demonstrate that nSMase2 plays a role in the production of Aß by stabilizing the interaction of APP with BACE1 in liquid ordered membrane microdomains, and directly protects neurons from the toxic effects of Aß. The effects of inhibiting nSMase2 on EV biogenesis may be independent from effects on Aß production and neuronal protection.


Asunto(s)
Enfermedad de Alzheimer , Ratones , Humanos , Animales , Secretasas de la Proteína Precursora del Amiloide , Ratones Transgénicos , Ácido Aspártico Endopeptidasas , Péptidos beta-Amiloides , Neuronas , Precursor de Proteína beta-Amiloide/genética , Presenilina-1 , Modelos Animales de Enfermedad , Esfingomielina Fosfodiesterasa/genética
4.
Pharmaceutics ; 14(10)2022 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-36297501

RESUMEN

Alzheimer's disease (AD) is characterized by the progressive accumulation of amyloid-ß and hyperphosphorylated tau (pTau), which can spread throughout the brain via extracellular vesicles (EVs). Membrane ceramide enrichment regulated by the enzyme neutral sphingomyelinase 2 (nSMase2) is a critical component of at least one EV biogenesis pathway. Our group recently identified 2,6-Dimethoxy-4-(5-Phenyl-4-Thiophen-2-yl-1H-Imidazol-2-yl)-Phenol (DPTIP), the most potent (30 nM) and selective inhibitor of nSMase2 reported to date. However, DPTIP exhibits poor oral pharmacokinetics (PK), modest brain penetration, and rapid clearance, limiting its clinical translation. To enhance its PK properties, we conjugated DPTIP to a hydroxyl-PAMAM dendrimer delivery system, creating dendrimer-DPTIP (D-DPTIP). In an acute brain injury model, orally administered D-DPTIP significantly reduced the intra-striatal IL-1ß-induced increase in plasma EVs up to 72 h post-dose, while oral DPTIP had a limited effect. In a mouse tau propagation model, where a mutant hTau (P301L/S320F) containing adeno-associated virus was unilaterally seeded into the hippocampus, oral D-DPTIP (dosed 3× weekly) significantly inhibited brain nSMase2 activity and blocked the spread of pTau to the contralateral hippocampus. These data demonstrate that dendrimer conjugation of DPTIP improves its PK properties, resulting in significant inhibition of EV propagation of pTau in mice. Dendrimer-based delivery of DPTIP has the potential to be an exciting new therapeutic for AD.

5.
J Med Chem ; 65(16): 11111-11125, 2022 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-35930706

RESUMEN

Extracellular vesicles (EVs) can carry pathological cargo and play an active role in disease progression. Neutral sphingomyelinase-2 (nSMase2) is a critical regulator of EV biogenesis, and its inhibition has shown protective effects in multiple disease states. 2,6-Dimethoxy-4-(5-phenyl-4-thiophen-2-yl-1H-imidazol-2-yl)phenol (DPTIP) is one of the most potent (IC50 = 30 nM) inhibitors of nSMase2 discovered to date. However, DPTIP exhibits poor oral pharmacokinetics (PK), limiting its clinical development. To overcome DPTIP's PK limitations, we synthesized a series of prodrugs by masking its phenolic hydroxyl group. When administered orally, the best prodrug (P18) with a 2',6'-diethyl-1,4'-bipiperidinyl promoiety exhibited >fourfold higher plasma (AUC0-t = 1047 pmol·h/mL) and brain exposures (AUC0-t = 247 pmol·h/g) versus DPTIP and a significant enhancement of DPTIP half-life (2 h vs ∼0.5 h). In a mouse model of acute brain injury, DPTIP released from P18 significantly inhibited IL-1ß-induced EV release into plasma and attenuated nSMase2 activity. These studies report the discovery of a DPTIP prodrug with potential for clinical translation.


Asunto(s)
Profármacos , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Esterasas , Ratones , Fenoles/farmacología , Profármacos/farmacocinética , Esfingomielina Fosfodiesterasa
6.
Biochem Pharmacol ; 194: 114796, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34678224

RESUMEN

Extracellular Vesicles (EVs) are implicated in the spread of pathogenic proteinsin a growing number of neurological diseases. Given this, there is rising interest in developing inhibitors of Neutral Sphingomyelinase 2 (nSMase2), an enzyme critical in EV biogenesis. Our group recently discovered phenyl(R)-(1-(3-(3,4-dimethoxyphenyl)-2,6-dimethylimidazo[1,2-b]pyridazin-8-yl)pyrrolidin-3-yl)carbamate (PDDC), the first potent, selective, orally-available, and brain-penetrable nSMase2 inhibitor, capable of dose-dependently reducing EVs release in vitro and in vivo. Herein, using multiplexed Surface Plasmon Resonance imaging (SPRi), we evaluated which brain cell-derived EVs were affected by PDDC following acute brain injury. Mice were fed PDDC-containing chow at doses which gave steady PDDC brain exposures exceeding its nSMase2 IC50. Mice were then administered an intra-striatal IL-1ß injection and two hours later plasma and brain were collected. IL-1ß injection significantly increased striatal nSMase2 activity which was completely normalized by PDDC. Using SPRi, we found that IL-1ß-induced injury selectively increased plasma levels of CD171 + and PLP1 + EVs; this EV increase was normalized by PDDC. In contrast, GLAST1 + EVs were unchanged by IL-1ß or PDDC. IL-1ß injection selectively increased EVs released from activated versus non-activated microglia, indicated by the CD11b+/IB4 + ratio. The increase in EVs from CD11b + microglia was dramatically attenuated with PDDC. Taken together, our data demonstrate that following acute injury, brain nSMase2 activity is elevated. EVs released from neurons, oligodendrocytes, and activated microglial are increased in plasma and inhibition of nSMase2 with PDDC reduced these IL-1ß-induced changes implicating nSMase2 inhibition as a therapeutic target for acute brain injury.


Asunto(s)
Lesiones Encefálicas/enzimología , Vesículas Extracelulares/enzimología , Microglía/enzimología , Neuronas/enzimología , Oligodendroglía/enzimología , Esfingomielina Fosfodiesterasa/metabolismo , Animales , Lesiones Encefálicas/tratamiento farmacológico , Carnitina/administración & dosificación , Carnitina/análogos & derivados , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/enzimología , Vesículas Extracelulares/efectos de los fármacos , Inyecciones Intraventriculares , Interleucina-1beta/administración & dosificación , Masculino , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Pirenos/administración & dosificación , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores
7.
Sci Adv ; 6(40)2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33008902

RESUMEN

Myelination requires a highly organized synthesis of multiple lipid species that regulate myelin curvature and compaction. For reasons that are not understood, central nervous system remyelinated axons often have thin myelin sheaths with a disorganized structure susceptible to secondary demyelination. We found that expression of the sphingomyelin hydrolase neutral sphingomyelinase 2 (nSMase2) during the differentiation of oligodendrocyte progenitor cells (OPCs) to myelinating oligodendrocytes changes their response to inflammatory cytokines. OPCs do not express nSMase2 and exhibit a protective/regenerative response to tumor necrosis factor-α and interleukin-1ß. Oligodendrocytes express nSMase2 and exhibit a stress response to cytokine challenge that includes an overproduction of ceramide, a sphingolipid that forms negative curvatures in membranes. Pharmacological inhibition or genetic deletion of nSMase2 in myelinating oligodendrocytes normalized the ceramide content of remyelinated fibers and increased thickness and compaction. These results suggest that inhibition of nSMase2 could improve the quality of myelin and stabilize structure.


Asunto(s)
Remielinización , Ceramidas/metabolismo , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Remielinización/fisiología , Esfingomielina Fosfodiesterasa/metabolismo
8.
J Extracell Vesicles ; 10(2): e12035, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33408815

RESUMEN

Chronic inflammation is thought to contribute to the early pathogenesis of Alzheimer's disease (AD). However, the precise mechanism by which inflammatory cytokines promote the formation and deposition of Aß remains unclear. Available data suggest that applications of inflammatory cytokines onto isolated neurons do not promote the formation of Aß, suggesting an indirect mechanism of action. Based on evidence astrocyte derived extracellular vesicles (astrocyte derived EVs) regulate neuronal functions, and data that inflammatory cytokines can modify the molecular cargo of astrocyte derived EVs, we sought to determine if IL-1ß promotes the formation of Aß indirectly through actions of astrocyte derived EVs on neurons. The production of Aß was increased when neurons were exposed to astrocyte derived EVs shed in response to IL-1ß (astrocyte derived EV-IL-1ß). The mechanism for this effect involved an enrichment of Casein kinase 1 (CK1) in astrocyte derived EV-IL-1ß. This astrocyte derived CK1 was delivered to neurons where it formed a complex with neuronal APC and GSK3 to inhibit the ß-catenin degradation. Stabilized ß-catenin translocated to the nucleus and bound to Hnrnpc gene at promoter regions. An increased cellular concentration of hnRNP C promoted the translation of APP by outcompeting the translational repressor fragile X mental retardation protein (FMRP) bound to APP mRNA. An increased amount of APP protein became co-localized with BACE1 in enlarged membrane microdomains concurrent with increased production of Aß. These findings identify a mechanism whereby inflammation promotes the formation of Aß through the actions of astrocyte derived EV-IL-1ß on neurons.


Asunto(s)
Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/metabolismo , Astrocitos/metabolismo , Quinasa de la Caseína I/metabolismo , Vesículas Extracelulares/metabolismo , Inflamación/patología , Interleucina-1beta/farmacología , Neuronas/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Amiloide/química , Amiloide/efectos de los fármacos , Precursor de Proteína beta-Amiloide/genética , Animales , Astrocitos/efectos de los fármacos , Astrocitos/inmunología , Estudios de Casos y Controles , Quinasa de la Caseína I/genética , Vesículas Extracelulares/efectos de los fármacos , Vesículas Extracelulares/inmunología , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Masculino , Persona de Mediana Edad , Neuronas/efectos de los fármacos , Neuronas/inmunología , Ratas , Ratas Sprague-Dawley
9.
Neuroinformatics ; 18(1): 163-179, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31273627

RESUMEN

Despite a multitude of commercially available multi-electrode array (MEA) systems that are each capable of rapid data acquisition from cultured neurons or slice cultures, there is a general lack of available analysis tools. These analysis gaps restrict the efficient extraction of meaningful physiological features from data sets, and limit interpretation of how experimental manipulations modify neural network activity. Here, we present the development of a user-friendly, publicly-available software called MEAnalyzer. This software contains several spike train analysis methods including relevant statistical calculations, periodicity analysis, functional connectivity analysis, and advanced data visualizations in a user-friendly graphical user interface that requires no coding from the user. Widespread availability of this user friendly and mathematically advanced program will stimulate and enhance the use of MEA technologies.


Asunto(s)
Potenciales de Acción/fisiología , Encéfalo/fisiología , Microelectrodos , Neuronas/fisiología , Programas Informáticos , Algoritmos , Animales , Electrofisiología/métodos
10.
Br J Pharmacol ; 176(19): 3857-3870, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31273753

RESUMEN

BACKGROUND AND PURPOSE: Extracellular vesicles (EVs) are constitutively shed from cells and released by various stimuli. Their protein and RNA cargo are modified by the stimulus, and in disease conditions can carry pathological cargo involved in disease progression. Neutral sphingomyelinase 2 (nSMase2) is a major regulator in at least one of several independent routes of EV biogenesis, and its inhibition is a promising new therapeutic approach for neurological disorders. Unfortunately, known inhibitors exhibit µM potency, poor physicochemical properties, and/or limited brain penetration. Here, we sought to identify a drug-like inhibitor of nSMase2. EXPERIMENTAL APPROACH: We conducted a human nSMase2 high throughput screen (>365,000 compounds). Selected hits were optimized focusing on potency, selectivity, metabolic stability, pharmacokinetics, and ability to inhibit EV release in vitro and in vivo. KEY RESULTS: We identified phenyl(R)-(1-(3-(3,4-dimethoxyphenyl)-2,6-dimethylimidazo[1,2-b]pyridazin-8-yl)pyrrolidin-3-yl)-carbamate (PDDC), a potent (pIC50  = 6.57) and selective non-competitive inhibitor of nSMase2. PDDC was metabolically stable, with excellent oral bioavailability (%F = 88) and brain penetration (AUCbrain /AUCplasma  = 0.60). PDDC dose-dependently (pEC50  = 5.5) inhibited release of astrocyte-derived extracellular vesicles (ADEV). In an in vivo inflammatory brain injury model, PDDC robustly inhibited ADEV release and the associated peripheral immunological response. A closely related inactive PDDC analogue was ineffective. CONCLUSION AND IMPLICATIONS: PDDC is a structurally novel, potent, orally available, and brain penetrant inhibitor of nSMase2. PDDC inhibited release of ADEVs in tissue culture and in vivo. PDDC is actively being tested in animal models of neurological disease and, along with closely related analogues, is being considered for clinical translation.


Asunto(s)
Encéfalo/efectos de los fármacos , Vesículas Extracelulares/efectos de los fármacos , Animales , Astrocitos/química , Astrocitos/metabolismo , Encéfalo/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Vesículas Extracelulares/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Masculino , Ratones , Microsomas Hepáticos/química , Microsomas Hepáticos/metabolismo , Estructura Molecular , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad
11.
Sci Rep ; 8(1): 17715, 2018 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-30531925

RESUMEN

Brain injury and inflammation induces a local release of extracellular vesicles (EVs) from astrocytes carrying proteins, RNAs, and microRNAs into the circulation. When these vesicles reach the liver, they stimulate the secretion of cytokines that mobilize peripheral immune cell infiltration into the brain, which can cause secondary tissue damage and impair recovery. Recent studies suggest that suppression of EV biosynthesis through neutral sphingomyelinase 2 (nSMase2) inhibition may represent a new therapeutic strategy. Unfortunately, currently available nSMase2 inhibitors exhibit low potency (IC50 ≥ 1 µM), poor solubility and/or limited brain penetration. Through a high throughput screening campaign of >365,000 compounds against human nSMase2 we identified 2,6-Dimethoxy-4-(5-Phenyl-4-Thiophen-2-yl-1H-Imidazol-2-yl)-Phenol (DPTIP), a potent (IC50 30 nM), selective, metabolically stable, and brain penetrable (AUCbrain/AUCplasma = 0.26) nSMase2 inhibitor. DPTIP dose-dependently inhibited EV release in primary astrocyte cultures. In a mouse model of brain injury conducted in GFAP-GFP mice, DPTIP potently (10 mg/kg IP) inhibited IL-1ß-induced astrocyte-derived EV release (51 ± 13%; p < 0.001). This inhibition led to a reduction of cytokine upregulation in liver and attenuation of the infiltration of immune cells into the brain (80 ± 23%; p < 0.01). A structurally similar but inactive analog had no effect in vitro or in vivo.


Asunto(s)
Astrocitos/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encefalitis/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores , Animales , Astrocitos/metabolismo , Encéfalo/metabolismo , Línea Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalitis/metabolismo , Vesículas Extracelulares/efectos de los fármacos , Vesículas Extracelulares/metabolismo , Células HEK293 , Humanos , Ratones , Ratas , Regulación hacia Arriba/efectos de los fármacos
12.
Exp Neurobiol ; 27(4): 287-298, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30181691

RESUMEN

Ischemic stroke and cerebral infarction triggered by the blockage of blood supply can cause damage to the brain via a complex series of pathological changes. Recently, diverse therapies have emerged as promising candidates for the treatment of stroke. These treatments exert therapeutic effects by acting on diverse target molecules and cells in different time windows from the acute to chronic phases. Here, using immunohistochemistry, we show pathophysiological changes in the brain microenvironment at the hyperacute (within 6 h), acute (1~3 days), subacute (7 days), and chronic (1 month) phases following ischemic injury. Ischemic injury in rats was induced by occluding the middle cerebral artery and was validated by magnetic resonance imaging. The progression of damage to the brain was evaluated by immunohistochemistry for NeuN+ neurons, GFAP+ astrocytes, and Iba1+ microglia, and by the emergence of the cell death-related molecules such as AIF, FAF1, and activated caspase-3. Our data regarding the spatial and temporal information on pathophysiological changes may warrant the investigation of the timing of administration of therapeutic treatments in preclinical studies with an animal model of stroke.

13.
Cell Death Dis ; 9(3): 363, 2018 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-29507357

RESUMEN

Astrocytes are known to be critical regulators of neuronal function. However, relatively few mediators of astrocyte to neuron communication have been identified. Recent advancements in the biology of extracellular vesicles have begun to implicate astrocyte derived extracellular vesicles (ADEV) as mediators of astrocyte to neuron communication, suggesting that alterations in the release and/or composition of ADEVs could influence gliotransmission. TNFα and IL-1ß are key mediators of glial activation and neuronal damage, but the effects of these cytokines on the release or molecular composition of ADEVs is unknown. We found that ADEVs released in response to IL-1ß (ADEV-IL-1ß) and TNFα (ADEV-TNFα) were enriched with miRNAs that target proteins involved in neurotrophin signaling. We confirmed that miR-125a-5p and miR-16-5p (both enriched in ADEV-IL-1ß and ADEV-TNFα) targeted NTKR3 and its downstream effector Bcl2. Downregulation of these targets in neurons was associated with reductions in dendritic growth, dendritic complexity, reduced spike rates, and burst activity. Molecular interference of miR-125a-5p and miR-16-5p prevented ADEV-IL-1ß from reducing dendritic complexity, spike, and burst rates. These findings suggest that astrocytes respond to inflammatory challenge by modifying the miRNA cargo of ADEVs to diminish the activity of target neurons by regulating the translational expression of proteins controlling programs essential for synaptic stability and neuronal excitability.


Asunto(s)
Astrocitos/metabolismo , Vesículas Extracelulares/metabolismo , Interleucina-1beta/farmacología , MicroARNs/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Neuronas/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/farmacología , Animales , Astrocitos/efectos de los fármacos , Secuencia de Bases , Dendritas/efectos de los fármacos , Dendritas/metabolismo , Vesículas Extracelulares/efectos de los fármacos , Células HEK293 , Humanos , MicroARNs/genética , Red Nerviosa/efectos de los fármacos , Red Nerviosa/metabolismo , Neuronas/efectos de los fármacos , Ratas Sprague-Dawley , Receptor trkC/metabolismo
14.
ACS Chem Neurosci ; 9(4): 809-816, 2018 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-29257872

RESUMEN

Insulin delivery to the brain has emerged as an important therapeutic target for cognitive disorders associated with abnormal brain energy metabolism. Although insulin is transported across the blood-brain barrier, peripheral routes of administration are problematic due to systemic effects of insulin on blood glucose. Intranasal (IN) administration is being investigated as an alternative route. We conducted a head-to-head comparison of subcutaneous (SC) and IN insulin, assessing plasma and brain pharmacokinetics and blood glucose levels in the mouse. SC insulin (2.4 IU) achieved therapeutically relevant concentrations in the brain (AUCbrain = 2537 h·µIU/mL) but dramatically increased plasma insulin (AUCplasma = 520 351 h·*µIU/mL), resulting in severe hypoglycemia and in some cases death. IN administration of the same dose resulted in similar insulin levels in the brain (AUCbrain = 3442 h·µIU/mL) but substantially lower plasma concentrations (AUCplasma = 354 h·µIU/mL), amounting to a ∼ 2000-fold increase in the AUCbrain:plasma ratio relative to SC. IN dosing also had no significant effect on blood glucose. When administered daily for 9 days, IN insulin increased brain glucose and energy metabolite concentrations (e.g., adenosine triphosphate and phosphocreatine) without causing overt toxicity, suggesting that IN insulin may be a safe therapeutic option for cognitively impaired patients.


Asunto(s)
Glucemia/metabolismo , Encéfalo/metabolismo , Insulina/sangre , Insulina/farmacocinética , Administración Intranasal , Animales , Barrera Hematoencefálica/metabolismo , Trastornos del Conocimiento/metabolismo , Metabolismo Energético/fisiología , Insulina/administración & dosificación , Insulina/líquido cefalorraquídeo , Masculino , Ratones
15.
Sci Rep ; 7(1): 7748, 2017 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-28798382

RESUMEN

The widespread use of combinational antiretroviral therapies (cART) in developed countries has changed the course of Human Immunodeficiency Virus (HIV) infection from an almost universally fatal disease to a chronic infection for the majority of individuals. Although cART has reduced the severity of neurological damage in HIV-infected individuals, the likelihood of cognitive impairment increases with age, and duration of infection. As cART does not suppress the expression of HIV non-structural proteins, it has been proposed that a constitutive production of HIV regulatory proteins in infected brain cells may contribute to neurological damage. However, this assumption has never been experimentally tested. Here we take advantage of the leaky tetracycline promoter system in the Tat-transgenic mouse to show that a chronic very low-level expression of Tat is associated with astrocyte activation, inflammatory cytokine expression, ceramide accumulation, reductions in brain volume, synaptic, and axonal damage that occurs over a time frame of 1 year. These data suggest that a chronic low-level production of Tat may contribute to progressive neurological damage in virally suppressed HIV-infected individuals.


Asunto(s)
Envejecimiento/patología , Encéfalo/virología , Enfermedades Neurodegenerativas/virología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Animales , Astrocitos/virología , Encéfalo/crecimiento & desarrollo , Ceramidas/metabolismo , Citocinas/metabolismo , Ratones , Fenotipo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
16.
Sci Signal ; 10(473)2017 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-28377412

RESUMEN

Brain injury induces a peripheral acute cytokine response that directs the transmigration of leukocytes into the brain. Because this brain-to-peripheral immune communication affects patient recovery, understanding its regulation is important. Using a mouse model of inflammatory brain injury, we set out to find a soluble mediator for this phenomenon. We found that extracellular vesicles (EVs) shed from astrocytes in response to intracerebral injection of interleukin-1ß (IL-1ß) rapidly entered into peripheral circulation and promoted the transmigration of leukocytes through modulation of the peripheral acute cytokine response. Bioinformatic analysis of the protein and microRNA cargo of EVs identified peroxisome proliferator-activated receptor α (PPARα) as a primary molecular target of astrocyte-shed EVs. We confirmed in mice that astrocytic EVs promoted the transmigration of leukocytes into the brain by inhibiting PPARα, resulting in the increase of nuclear factor κB (NF-κB) activity that triggered the production of cytokines in liver. These findings expand our understanding of the mechanisms regulating communication between the brain and peripheral immune system and identify astrocytic EVs as a molecular regulator of the immunological response to inflammatory brain damage.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/metabolismo , Vesículas Extracelulares/metabolismo , Mediadores de Inflamación/metabolismo , Leucocitos Mononucleares/metabolismo , Animales , Animales Recién Nacidos , Western Blotting , Encéfalo/efectos de los fármacos , Encéfalo/patología , Células Cultivadas , Ceramidas/metabolismo , Citocinas/genética , Citocinas/metabolismo , Vesículas Extracelulares/ultraestructura , Interleucina-1beta/farmacología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Electrónica , Microscopía Fluorescente , Interferencia de ARN , Esfingomielina Fosfodiesterasa/genética , Esfingomielina Fosfodiesterasa/metabolismo , Migración Transcelular de la Célula/efectos de los fármacos , Migración Transcelular de la Célula/genética
17.
J Biol Chem ; 292(7): 2557-2570, 2017 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-28049733

RESUMEN

All vertebrate cell surfaces display a dense glycan layer often terminated with sialic acids, which have multiple functions due to their location and diverse modifications. The major sialic acids in most mammalian tissues are N-acetylneuraminic acid (Neu5Ac) and N-glycolylneuraminic acid (Neu5Gc), the latter being derived from Neu5Ac via addition of one oxygen atom at the sugar nucleotide level by CMP-Neu5Ac hydroxylase (Cmah). Contrasting with other organs that express various ratios of Neu5Ac and Neu5Gc depending on the variable expression of Cmah, Neu5Gc expression in the brain is extremely low in all vertebrates studied to date, suggesting that neural expression is detrimental to animals. However, physiological exploration of the reasons for this long term evolutionary selection has been lacking. To explore the consequences of forced expression of Neu5Gc in the brain, we have established brain-specific Cmah transgenic mice. Such Neu5Gc overexpression in the brain resulted in abnormal locomotor activity, impaired object recognition memory, and abnormal axon myelination. Brain-specific Cmah transgenic mice were also lethally sensitive to a Neu5Gc-preferring bacterial toxin, even though Neu5Gc was overexpressed only in the brain and other organs maintained endogenous Neu5Gc expression, as in wild-type mice. Therefore, the unusually strict evolutionary suppression of Neu5Gc expression in the vertebrate brain may be explained by evasion of negative effects on neural functions and by selection against pathogens.


Asunto(s)
Evolución Biológica , Encéfalo/metabolismo , Ácidos Neuramínicos/metabolismo , Animales , Cromatografía Líquida de Alta Presión , Endotelio Vascular/metabolismo , Locomoción , Espectrometría de Masas , Trastornos de la Memoria/metabolismo , Ratones , Ratones Transgénicos
18.
J Comp Neurol ; 525(2): 291-301, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27339102

RESUMEN

The corpus callosum (CC) is the largest fiber tract in the mammalian brain, linking the bilateral cerebral hemispheres. CC development depends on the proper balance of axon growth cone attractive and repellent cues leading axons to the midline and then directing them to the contralateral hemisphere. Imbalance of these cues results in CC agenesis or dysgenesis. Nogo receptors (NgR1, NgR2, and NgR3) are growth cone directive molecules known for inhibiting axon regeneration after injury. We report that mice lacking Nogo receptors (NgR123-null mice) display complete CC agenesis due to axon misdirection evidenced by ectopic axons including cortical Probst bundles. Because glia and glial-derived growth cone repellent factors (especially the diffusible factor Slit2) are required for CC development, their distribution was studied. Compared with wild-type mice, NgR123-null mice had a sharp increase in the glial marker glial fibrillary acidic protein (GFAP) and in Slit2 at the glial wedge and indusium griseum, midline structures required for CC formation. NgR123-null mice displayed reduced motor coordination and hyperactivity. These data are consistent with the hypotheses that Nogo receptors are membrane-bound growth cone repellent factors required for migration of axons across the midline at the CC, and that their absence results directly or indirectly in midline gliosis, increased Slit2, and complete CC agenesis. J. Comp. Neurol. 525:291-301, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Agenesia del Cuerpo Calloso/genética , Cuerpo Calloso/embriología , Neurogénesis/fisiología , Receptores Nogo/deficiencia , Animales , Modelos Animales de Enfermedad , Immunoblotting , Inmunohistoquímica , Ratones , Ratones Noqueados
19.
Ann Rehabil Med ; 40(6): 961-971, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28119825

RESUMEN

OBJECTIVE: To determine the efficacy of a stretching and strengthening exercise program using an upper extremity robot, as compared with a conventional occupational therapy program for upper extremity spasticity in stroke patients. METHODS: Subjects were randomly divided into a robot-assisted therapy (RT) group and a conventional rehabilitation therapy (CT) group. RT group patients received RT and CT once daily for 30 minutes each, 5 days a week, for 2 weeks. RT was performed using an upper-extremity robot (Neuro-X; Apsun Inc., Seoul, Korea), and CT was administered by occupational therapists. CT group patients received CT alone twice daily for 30 minutes, 5 days a week, for 2 weeks. Modified Ashworth Scale (MAS) was used to measure the spasticity of upper extremity. Manual muscle tests (MMT), Manual Function Tests (MFT), Brunnstrom stage, and the Korean version of Modified Barthel Index (K-MBI) were used to measure the strength and function of upper extremity. All measurements were obtained before and after 2-week treatment. RESULTS: The RT and CT groups included 22 subjects each. After treatment, both groups showed significantly lower MAS scores and significant improvement in the MMT, MFT, Brunnstrom stage, and K-MBI scores. Treatment effects showed no significant differences between the two groups. CONCLUSION: RT showed similar treatment benefits on spasticity, as compared to CT. The study results suggested that RT could be a useful method for continuous, repeatable, and relatively accurate range of motion exercise in stroke patients with spasticity.

20.
PLoS One ; 10(5): e0124481, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26010541

RESUMEN

Ceramide is a bioactive lipid that plays an important role in stress responses leading to apoptosis, cell growth arrest and differentiation. Ceramide production is due in part to sphingomyelin hydrolysis by sphingomyelinases. In brain, neutral sphingomyelinase 2 (nSMase2) is expressed in neurons and increases in its activity and expression have been associated with pro-inflammatory conditions observed in Alzheimer's disease, multiple sclerosis and human immunodeficiency virus (HIV-1) patients. Increased nSMase2 activity translates into higher ceramide levels and neuronal cell death, which can be prevented by chemical or genetic inhibition of nSMase2 activity or expression. However, to date, there are no soluble, specific and potent small molecule inhibitor tool compounds for in vivo studies or as a starting point for medicinal chemistry optimization. Moreover, the majority of the known inhibitors were identified using bacterial, bovine or rat nSMase2. In an attempt to identify new inhibitor scaffolds, two activity assays were optimized as screening platform using the recombinant human enzyme. First, active hits were identified using a fluorescence-based high throughput compatible assay. Then, hits were confirmed using a 14C sphingomyelin-based direct activity assay. Pharmacologically active compounds and approved drugs were screened using this strategy which led to the identification of cambinol as a novel uncompetitive nSMase2 inhibitor (Ki = 7 µM). The inhibitory activity of cambinol for nSMase2 was approximately 10-fold more potent than for its previously known target, silence information regulator 1 and 2 (SIRT1/2). Cambinol decreased tumor necrosis factor-α or interleukin-1 ß-induced increases of ceramide and cell death in primary neurons. A preliminary study of cambinol structure and activity allowed the identification of the main structural features required for nSMase2 inhibition. Cambinol and its analogs may be useful as nSMase2 inhibitor tool compounds to prevent ceramide-dependent neurodegeneration.


Asunto(s)
Naftalenos/farmacología , Fármacos Neuroprotectores/farmacología , Pirimidinonas/farmacología , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores , Animales , Bovinos , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ceramidas/biosíntesis , Citocinas/farmacología , Dendritas/efectos de los fármacos , Dendritas/patología , Evaluación Preclínica de Medicamentos , Pruebas de Enzimas , Inhibidores Enzimáticos/farmacología , Fluorescencia , Células HEK293 , Hipocampo/patología , Humanos , Interleucina-1beta/farmacología , Naftalenos/química , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/química , Pirimidinonas/química , Radiactividad , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacología , Esfingomielina Fosfodiesterasa/metabolismo , Relación Estructura-Actividad , Factor de Necrosis Tumoral alfa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...