Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Neuroinflammation ; 19(1): 315, 2022 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-36577999

RESUMEN

BACKGROUND: Dysregulated activation of the inflammasome is involved in various human diseases including acute cerebral ischemia, multiple sclerosis and sepsis. Though many inflammasome inhibitors targeting NOD-like receptor protein 3 (NLRP3) have been designed and developed, none of the inhibitors are clinically available. Growing evidence suggests that targeting apoptosis-associated speck-like protein containing a CARD (ASC), the oligomerization of which is the key event for the assembly of inflammasome, may be another promising therapeutic strategy. Lonidamine (LND), a small-molecule inhibitor of glycolysis used as an antineoplastic drug, has been evidenced to have anti-inflammation effects. However, its anti-inflammatory mechanism is still largely unknown. METHODS: Middle cerebral artery occlusion (MCAO), experimental autoimmune encephalomyelitis (EAE) and LPS-induced sepsis mice models were constructed to investigate the therapeutic and anti-inflammasome effects of LND. The inhibition of inflammasome activation and ASC oligomerization by LND was evaluated using western blot (WB), immunofluorescence (IF), quantitative polymerase chain reaction (qPCR) and enzyme-linked immunosorbent assay (ELISA) in murine bone marrow-derived macrophages (BMDMs). Direct binding of LND with ASC was assessed using molecular mock docking, surface plasmon resonance (SPR), and drug affinity responsive target stability (DARTS). RESULTS: Here, we find that LND strongly attenuates the inflammatory injury in experimental models of inflammasome-associated diseases including autoimmune disease-multiple sclerosis (MS), ischemic stroke and sepsis. Moreover, LND blocks diverse types of inflammasome activation independent of its known targets including hexokinase 2 (HK2). We further reveal that LND directly binds to the inflammasome ligand ASC and inhibits its oligomerization. CONCLUSIONS: Taken together, our results identify LND as a broad-spectrum inflammasome inhibitor by directly targeting ASC, providing a novel candidate drug for the treatment of inflammasome-driven diseases in clinic.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Sepsis , Humanos , Ratones , Animales , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico
2.
Mar Drugs ; 17(5)2019 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-31060323

RESUMEN

High intraocular pressure (IOP)-induced retinal ischemia leads to acute glaucoma, which is one of the leading causes of irreversible visual-field loss, characterized by loss of retinal ganglion cells (RGCs) and axonal injury in optic nerves (ONs). Oxidative stress and the inflammatory response play an important role in the ischemic injury of retinal and optic nerves. We focus on 5α-androst-3ß, 5α, 6ß-triol (TRIOL), a synthetic neuroactive derivative of natural marine steroids 24-methylene-cholest-3ß, 5α, 6ß, 19-tetrol and cholestane-3ß, 5α, 6ß-triol, which are two neuroactive polyhydroxysterols isolated from the soft coral Nephthea brassica and the gorgonian Menella kanisa, respectively. We previously demonstrated that TRIOL was a neuroprotective steroid with anti-inflammatory and antioxidative activities. However, the potential role of TRIOL on acute glaucoma and its underlying mechanisms remains unclear. Here, we report TRIOL as a promising neuroprotectant that can protect RGCs and their axons/dendrites from ischemic-reperfusion (I/R) injury in an acute intraocular hypertension (AIH) model. Intravitreal injection of TRIOL significantly alleviated the loss of RGCs and the damage of axons and dendrites in rats and mice with acute glaucoma. As NF-E2-related factor 2 (Nrf2) is one of the most critical regulators in oxidative and inflammatory injury, we further evaluated the effect of TRIOL on Nrf2 knockout mice, and the neuroprotective role of TRIOL on retinal ischemia was not observed in Nrf2 knockout mice, indicating that activation of Nrf2 is responsible for the neuroprotection of TRIOL. Further experiments demonstrated that TRIOL can activate and upregulate Nrf2, along with its downstream hemeoxygenase-1 (HO-1), by negative regulation of Kelch-like ECH (Enoyl-CoA Hydratase) associated Protein-1 (Keap1). In conclusion, our study shed new light on the neuroprotective therapy of retinal ischemia and proposed a promising marine drug candidate, TRIOL, for the therapeutics of acute glaucoma.


Asunto(s)
Androstanoles/farmacología , Factor 2 Relacionado con NF-E2/deficiencia , Fármacos Neuroprotectores/farmacología , Daño por Reperfusión/tratamiento farmacológico , Células Ganglionares de la Retina/efectos de los fármacos , Esteroides/farmacología , Animales , Técnicas de Cultivo de Célula , Hipoxia de la Célula/efectos de los fármacos , Modelos Animales de Enfermedad , Glaucoma , Hemo-Oxigenasa 1/metabolismo , Inflamación/tratamiento farmacológico , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/efectos de los fármacos , Microglía/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Hipertensión Ocular/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
3.
Molecules ; 24(2)2019 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-30641903

RESUMEN

Neuroinflammation has been well recognized as a key pathological event in acute glaucoma. The medical therapy of acute glaucoma mainly focuses on lowering intraocular pressure (IOP), while there are still scarce anti-inflammatory agents in the clinical treatment of acute glaucoma. Here we reported that ß,3α,5α-trihydroxy-androst-6-one (sterone), a novel synthetic polyhydric steroid, blocked neuroinflammation mediated by microglia/macrophages and alleviated the loss of retinal ganglion cells (RGCs) caused by acute intraocular hypertension (AIH). The results showed that sterone significantly inhibited the morphological changes, the up-regulation of inflammatory biomarker ionized calcium-binding adapter molecule 1 (Iba-1), and the mRNA increase of proinflammatory tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), and interleukin-6 (IL-6) induced by lipopolysaccharide (LPS) in BV2 microglia and RAW264.7 macrophages. Moreover, immunofluorescence and western blotting analysis revealed that sterone markedly abrogated the nuclear translocation and phosphorylation of nuclear factor-κB (NF-κB) p65 subunit. Furthermore, sterone significantly suppressed the inflammatory microglial activation and RGCs' reduction caused by retinal ischemia/reperfusion (I/R) injury in a rat AIH model. These results suggest sterone may be a potential candidate in the treatment of acute glaucoma caused by microglial activation-mediated neuroinflammatory injury.


Asunto(s)
Microglía/efectos de los fármacos , Microglía/metabolismo , Fármacos Neuroprotectores/farmacología , Hipertensión Ocular/metabolismo , Hipertensión Ocular/fisiopatología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Esteroides/farmacología , Enfermedad Aguda , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacología , Recuento de Células , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Glaucoma/tratamiento farmacológico , Glaucoma/etiología , Glaucoma/metabolismo , Glaucoma/fisiopatología , Lipopolisacáridos/efectos adversos , Ratones , Estructura Molecular , FN-kappa B/metabolismo , Fármacos Neuroprotectores/síntesis química , Hipertensión Ocular/tratamiento farmacológico , Hipertensión Ocular/etiología , Células RAW 264.7 , Ratas , Daño por Reperfusión/etiología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Esteroides/síntesis química
4.
J Neurochem ; 144(2): 186-200, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29205357

RESUMEN

Hyperglycolysis, observed within the penumbra zone during brain ischemia, was shown to be detrimental for tissue survival because of lactate accumulation and reactive oxygen species overproduction in clinical and experimental settings. Recently, mounting evidence suggests that glycolytic reprogramming and induced metabolic enzymes can fuel the activation of peripheral immune cells. However, the possible roles and details regarding hyperglycolysis in neuroinflammation during ischemia are relatively poorly understood. Here, we investigated whether overactivated glycolysis could activate microglia and identified the crucial regulators of neuroinflammatory responses in vitro and in vivo. Using BV 2 and primary microglial cultures, we found hyperglycolysis and induction of the key glycolytic enzyme hexokinase 2 (HK2) were essential for microglia-mediated neuroinflammation under hypoxia. Mechanistically, HK2 up-regulation led to accumulated acetyl-coenzyme A, which accounted for the subsequent histone acetylation and transcriptional activation of interleukin (IL)-1ß. The inhibition and selective knockdown of HK2 in vivo significantly protected against ischemic brain injury by suppressing microglial activation and IL-1ß production in male Sprague-Dawley rats subjected to transient middle cerebral artery occlusion (MCAo) surgery. We provide novel insights for HK2 specifically serving as a neuroinflammatory determinant, thus explaining the neurotoxic effect of hyperglycolysis and indicating the possibility of selectively targeting HK2 as a therapeutic strategy in acute ischemic stroke.


Asunto(s)
Isquemia Encefálica/enzimología , Isquemia Encefálica/genética , Glucólisis/genética , Hexoquinasa/genética , Hexoquinasa/metabolismo , Activación de Macrófagos/genética , Microglía/enzimología , Accidente Cerebrovascular/enzimología , Accidente Cerebrovascular/genética , Acetilcoenzima A/metabolismo , Acetilación , Animales , Inducción Enzimática/genética , Hexoquinasa/biosíntesis , Histonas/metabolismo , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/metabolismo , Inflamación/genética , Interleucina-1beta/metabolismo , Masculino , Interferencia de ARN , Ratas , Ratas Sprague-Dawley
5.
Biochem Biophys Res Commun ; 496(1): 95-100, 2018 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-29307820

RESUMEN

Neuronal hyperexcitability is identified as a critical pathological basis of epileptic seizures. Cholestane-3ß, 5α, 6ß-triol (Triol) is a major metabolic oxysterol of cholesterol. Although its neuroprotective effect on ischemia-induced neuronal injury and negative modulation of voltage-gated sodium (Nav) channels were well established, the physical binding site of triol to sodium channels and its effects on neuronal hyperexcitability have not yet been explored. In this study, we utilized molecular docking and molecular dynamics simulation to investigate the interaction between triol and Nav Channels. Our results demonstrated that triol binds to the indole ring of Trp122 of the Nav Channel in silico with a high biological affinity. We further found that triol negatively modulates the action potentials bursts of hippocampal neurons by cell-attached patch recording. Moreover, triol significantly inhibits low Mg2+-induced hyperexcitability in vitro. In addition, triol attenuates pentylenetetrazole (PTZ)-induced convulsive-form behavioral deficits in vivo. Together, our results suggest that triol suppresses neuronal hyperexcitability via binding to Nav channel, indicating that triol might be an attractive lead compound for the treatment of neuronal hyperexcitability-related neurological disorders, especially epileptic seizures.


Asunto(s)
Potenciales de Acción/fisiología , Colestanoles/administración & dosificación , Colestanoles/química , Epilepsia/prevención & control , Neuronas/fisiología , Canales de Sodio Activados por Voltaje/química , Canales de Sodio Activados por Voltaje/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Sitios de Unión , Células Cultivadas , Relación Dosis-Respuesta a Droga , Epilepsia/fisiopatología , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Unión Proteica , Conformación Proteica , Ratas , Ratas Sprague-Dawley , Resultado del Tratamiento
6.
Biochem Biophys Res Commun ; 483(2): 892-896, 2017 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-28082198

RESUMEN

Neuroinflammation is one of key pathologic element in neurological diseases including stroke, traumatic brain injury, Alzheimer' s Disease, Parkinson's Disease, and multiple sclerosis as well. Up-regulation of endothelial adhesion molecules, which facilitate leukocyte adhesion to the endothelium, is the vital process of endothelial cells mediated neuroinflammation. Androst-3ß, 5α, 6ß-triol (Triol) is a synthetic steroid which has been reported to have neuroprotective effects in hypoxia/re-oxygenation-induced neuronal injury model. In the present study, we firstly investigated whether Triol inhibited the TNF-α-induced inflammatory response in rat brain microvascular endothelial cells (RBMECs). Our data showed that Triol decreased TNF-α-induced expression of vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1) and the adhesion of neutrophil to RBMECs. We also found that Triol inhibited TNF-α-induced degradation of IκBα and phosphorylation of NF-κBp65 that are required for NF-κB activation. Furthermore, Triol significantly reversed TNF-α-induced down-expression of CYLD, which is a deubiquitinase that negatively regulates activation of NF-κB. These results suggest that Triol displays an anti-inflammatory effect on TNF-α-induced RBMECs via downregulating of CYLD-NF-κB signaling pathways and might have a potential benefit in therapeutic neuroinflammation related diseases.


Asunto(s)
Androstanoles/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Fármacos Neuroprotectores/farmacología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Animales , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Células Endoteliales/citología , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Inhibidor NF-kappaB alfa/metabolismo , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Fosforilación , Ratas , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Ubiquitina Tiolesterasa/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
7.
ACS Chem Neurosci ; 13(14): 2110-2121, 2022 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-35770894

RESUMEN

As one of the key injury incidents, tissue acidosis in the brain occurs very quickly within several minutes upon the onset of ischemic stroke. Glutamate, an excitatory amino acid inducing neuronal excitotoxicity, has been reported to trigger the decrease in neuronal intracellular pH (pHi) via modulating proton-related membrane transporters. However, there remains a lack of clarity on the possible role of glutamate in neuronal acidosis via regulating metabolism. Here, we show that 200 µM glutamate treatment quickly promotes glycolysis and inhibits mitochondrial oxidative phosphorylation of primary cultured neurons within 15 min, leading to significant cytosolic lactate accumulation, which contributes to the rapid intracellular acidification and neuronal injury. The reprogramming of neuronal metabolism by glutamate is dependent on adenosine monophosphate-activated protein kinase (AMPK) signaling since the inhibition of AMPK activation by its selective inhibitor compound C significantly reverses these deleterious events in vitro. Moreover, 5α-androst-3ß,5α,6ß-TRIOL (TRIOL), a neuroprotectant we previously reported, can also remarkably reverse intracellular acidification and alleviate neuronal injury through the inhibition of AMPK signaling. Furthermore, TRIOL remarkably reduced the infarct volume and attenuated neurologic impairment in acute ischemic stroke models of middle cerebral artery occlusion in vivo. In summary, we reveal a novel role of glutamate in rapid intracellular acidification injury resulting from glutamate-induced lactate accumulation through AMPK-mediated neuronal reprogramming. Moreover, inhibition of the quick drop in neuronal pHi by TRIOL significantly reduces the cerebral damages, suggesting that it is a promising drug candidate for ischemic stroke.


Asunto(s)
Lesiones Encefálicas , Accidente Cerebrovascular Isquémico , Proteínas Quinasas Activadas por AMP , Ácido Glutámico , Humanos , Concentración de Iones de Hidrógeno , Lactatos , Neuronas/fisiología , Fármacos Neuroprotectores
8.
Nat Med ; 10(7): 719-26, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15195085

RESUMEN

We have previously shown that apolipoprotein E (Apoe) promotes the formation of amyloid in brain and that astrocyte-specific expression of APOE markedly affects the deposition of amyloid-beta peptides (Abeta) in a mouse model of Alzheimer disease. Given the capacity of astrocytes to degrade Abeta, we investigated the potential role of Apoe in this astrocyte-mediated degradation. In contrast to cultured adult wild-type mouse astrocytes, adult Apoe(-/-) astrocytes do not degrade Abeta present in Abeta plaque-bearing brain sections in vitro. Coincubation with antibodies to either Apoe or Abeta, or with RAP, an antagonist of the low-density lipoprotein receptor family, effectively blocks Abeta degradation by astrocytes. Phase-contrast and confocal microscopy show that Apoe(-/-) astrocytes do not respond to or internalize Abeta deposits to the same extent as do wild-type astrocytes. Thus, Apoe seems to be important in the degradation and clearance of deposited Abeta species by astrocytes, a process that may be impaired in Alzheimer disease.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Apolipoproteínas E/fisiología , Astrocitos/metabolismo , Animales , Agregación Celular , Supervivencia Celular , Células Cultivadas , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/fisiología , Ratones , Ratones Endogámicos C57BL
9.
Zool Res ; 42(2): 250-251, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33738990

RESUMEN

Following the publication of our paper (Zhang et al., 2020), it has come to our attention that we erroneously listed two funding sources unrelated to this study in the "ACKNOWLEDGEMENTS" section. Hereby, we wish to update the "ACKNOWLEDGEMENTS" section as a correction.

10.
J Neurosci ; 29(21): 6771-9, 2009 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-19474305

RESUMEN

To investigate the role of human apolipoprotein E (apoE) on Abeta deposition in vivo, we crossed PDAPP mice lacking mouse Apoe to targeted replacement mice expressing human apoE (PDAPP/TRE2, PDAPP/TRE3, or PDAPP/TRE4). We then measured the levels of apoE protein and Abeta peptides in plasma, CSF, and brain homogenates in these mice at different ages. We also quantified the amount of brain Abeta and amyloid burden in 18-month-old mice. In young PDAPP/TRE4 mice that were analyzed at an age before brain Abeta deposition, we observed a significant decrease in the levels of apoE in CSF and brain when compared with age-matched mice expressing either human E2 or E3. The brain levels of Abeta42 in PDAPP/TRE4 mice were substantially elevated even at this very early time point. In older PDAPP/TRE4 mice, the levels of insoluble apoE protein increased in parallel to the dramatic rise in brain Abeta burden, and the majority of apoE was associated with Abeta. In TRE4 only mice, we also observed a significant decrease in the level of apoE in brain homogenates. Since the relative level of apoE mRNA was equivalent in PDAPP/TRE and TRE only mice, it appears that post-translational mechanisms influence the levels of apoE protein in brain (E4 < E3 << E2), resulting in early and dramatic apoE isoform-dependent effects on brain Abeta levels (E4 >> E3 > E2) that increase with age. Therapeutic strategies aimed at increasing the soluble levels of apoE protein, regardless of isoform, may effectively prevent and (or) treat Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Apolipoproteínas E/metabolismo , Encéfalo/metabolismo , Factores de Edad , Análisis de Varianza , Animales , Apolipoproteínas E/clasificación , Apolipoproteínas E/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Fragmentos de Péptidos/metabolismo , Isoformas de Proteínas/metabolismo
11.
J Neurosci ; 29(11): 3603-12, 2009 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-19295164

RESUMEN

Recent studies suggest that bone marrow-derived macrophages can effectively reduce beta-amyloid (Abeta) deposition in brain. To further elucidate the mechanisms by which macrophages degrade Abeta, we cultured murine macrophages on top of Abeta plaque-bearing brain sections from transgenic mice expressing PDAPP [human amyloid precursor protein (APP) with the APP(717V>F) mutation driven by the platelet-derived growth factor promoter]. Using this ex vivo assay, we found that macrophages from wild-type mice very efficiently degrade both soluble and insoluble Abeta in a time-dependent manner and markedly eliminate thioflavine-S positive amyloid deposits. Because macrophages express and secrete apolipoprotein E (apoE), we compared the efficiency of Abeta degradation by macrophages prepared from apoE-deficient mice or mice expressing human apoE2, apoE3, or apoE4. Macrophages expressing apoE2 were more efficient at degrading Abeta than apoE3-expressing, apoE4-expressing, or apoE-deficient macrophages. Moreover, macrophage-induced degradation of Abeta was effectively blocked by an anti-apoE antibody and receptor-associated protein, an antagonist of the low-density lipoprotein (LDL) receptor family, suggesting involvement of LDL receptors. Measurement of matrix metalloproteinase-9 (MMP-9) activity in the media from human apoE-expressing macrophages cocultured with Abeta-containing brain sections revealed greater levels of MMP-9 activity in apoE2-expressing than in either apoE3- or apoE4-expressing macrophages. Differences in MMP-9 activity appear to contribute to the isoform-specific differences in Abeta degradation by macrophages. These apoE isoform-dependent effects of macrophages on Abeta degradation suggest a novel "peripheral" mechanism for Abeta clearance from brain that may also, in part, explain the isoform-dependent effects of apoE in determining the genetic risk for Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Amiloide/metabolismo , Apolipoproteínas E/fisiología , Macrófagos/fisiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/biosíntesis , Precursor de Proteína beta-Amiloide/genética , Animales , Apolipoproteínas E/genética , Células Cultivadas , Técnicas de Cocultivo/métodos , Humanos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología
12.
Oxid Med Cell Longev ; 2020: 2048210, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33163142

RESUMEN

Nuclear receptor subfamily 4 group A member 3 (NR4A3) protects the vascular endothelial cell (VEC) against hypoxia stress, whose expression is primarily reported to be governed at a transcriptional level. However, the regulation of NR4A3 in the protein level is largely unknown. Here, we report that NR4A3 protein abundance is decreased immensely in VEC injury induced by reoxygenation after oxygen-glucose deprivation (OGD-R), which is significantly blocked by the administration of the antioxidative steroid TRIOL. Moreover, the notable improvement of NR4A3 and the alleviation of pulmonary endothelial barrier hyperpermeability induced by acute hypobaric hypoxia in cynomolgus monkeys are also observed after TRIOL administration. The overproduction of reactive oxygen species (ROS) decreases NR4A3 protein abundance in VEC under OGD-R condition, which is reversed by TRIOL and N-acetylcysteine (NAC). TRIOL dose-dependently increases the NR4A3 protein level by inhibiting ubiquitination and ubiquitin proteasome system- (UPS-) mediated degradation rather than promoting its transcription. Using yeast two-hybrid screening, we further identify the interaction between NR4A3 and SWI/SNF-related matrix-associated actin-dependent regulator of chromatin subfamily B member 1 (SMARCB1), and the DNA-binding domain of NR4A3 is required for this interaction. Knockdown of SMARCB1 reduces ubiquitination and degradation of NR4A3, suggesting the proubiquitylation effect of this interaction which is enhanced by ROS in VEC injury induced by OGD-R. In summary, our study here for the first time reveals a posttranslational regulation in SMARCB1-mediated NR4A3 protein degradation which is driven by ROS, providing further understanding of the impaired regulation of NR4A3-mediated prosurvival pathways under pathological condition in VEC.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Endotelio Vascular/lesiones , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Hipoxia/metabolismo , Pulmón/metabolismo , Proteolisis , Especies Reactivas de Oxígeno , Receptores de Esteroides/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Proteína SMARCB1/metabolismo , Ubiquitinación , Animales , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Pulmón/patología , Macaca fascicularis
13.
Zool Res ; 41(1): 3-19, 2020 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-31840949

RESUMEN

Hypobaric hypoxia (HH) exposure can cause serious brain injury as well as life-threatening cerebral edema in severe cases. Previous studies on the mechanisms of HH-induced brain injury have been conducted primarily using non-primate animal models that are genetically distant to humans, thus hindering the development of disease treatment. Here, we report that cynomolgus monkeys ( Macacafascicularis) exposed to acute HH developed human-like HH syndrome involving severe brain injury and abnormal behavior. Transcriptome profiling of white blood cells and brain tissue from monkeys exposed to increasing altitude revealed the central role of the HIF-1 and other novel signaling pathways, such as the vitamin D receptor (VDR) signaling pathway, in co-regulating HH-induced inflammation processes. We also observed profound transcriptomic alterations in brains after exposure to acute HH, including the activation of angiogenesis and impairment of aerobic respiration and protein folding processes, which likely underlie the pathological effects of HH-induced brain injury. Administration of progesterone (PROG) and steroid neuroprotectant 5α-androst-3ß,5,6ß-triol (TRIOL) significantly attenuated brain injuries and rescued the transcriptomic changes induced by acute HH. Functional investigation of the affected genes suggested that these two neuroprotectants protect the brain by targeting different pathways, with PROG enhancing erythropoiesis and TRIOL suppressing glutamate-induced excitotoxicity. Thus, this study advances our understanding of the pathology induced by acute HH and provides potential compounds for the development of neuroprotectant drugs for therapeutic treatment.


Asunto(s)
Androstanoles/farmacología , Hipoxia/veterinaria , Macaca fascicularis , Enfermedades de los Monos/prevención & control , Progesterona/farmacología , Transcriptoma , Androstanoles/administración & dosificación , Animales , Encefalopatías/prevención & control , Encefalopatías/veterinaria , Calcio/metabolismo , Regulación de la Expresión Génica , Hipoxia/patología , Leucocitos/metabolismo , Masculino , Fármacos Neuroprotectores/farmacología , Presión , Progesterona/administración & dosificación
14.
FASEB J ; 22(1): 47-54, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17666454

RESUMEN

Cleavage of the amyloid precursor protein (APP) by beta- and gamma-secretases results in generation of the amyloid-beta protein (Abeta), which is characteristically deposited in the brain of Alzheimer's disease patients. Inhibitors of 3-hydroxy-3-methyl-glutaryl (HMG)-CoA reductase (the statins) reduce levels of cholesterol and isoprenoids such as geranylgeranyl pyrophosphate (GGPP). Previous studies have demonstrated that cholesterol increases and statins reduce Abeta levels mostly by regulating beta-secretase activity. In this study, we focused on the role of geranylgeranyl isoprenoids GGPP and geranylgeraniol (GGOH) in regulating Abeta production. Our data show that the inhibition of GGPP synthesis by statins plays an important role in statin-mediated reduction of Abeta secretion. Consistent with this finding, the geranylgeranyl isoprenoids preferentially increase the yield of Abeta of 42 residues (Abeta42) in a dose-dependent manner. Our studies further demonstrated that geranylgeranyl isoprenoids increase the yield of APP-CTFgamma (a.k.a. AICD) as well as Abeta by stimulating gamma-secretase-mediated cleavage of APP-CTFalpha and APP-CTFbeta in vitro. Furthermore, GGOH increases the levels of the active gamma-secretase complex in the detergent-insoluble membrane fraction along with its substrates, APP-CTFalpha and APP-CTFbeta. Our results indicate that geranylgeranyl isoprenoids may be an important physiological facilitator of gamma-secretase activity that can foster production of the pathologically important Abeta42.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/biosíntesis , Precursor de Proteína beta-Amiloide/biosíntesis , Fragmentos de Péptidos/biosíntesis , Fosfatos de Poliisoprenilo/farmacología , Secuencia de Aminoácidos , Animales , Células CHO , Línea Celular , Cricetinae , Cricetulus , Activación Enzimática , Ensayo de Inmunoadsorción Enzimática , Humanos , Lovastatina/farmacología , Datos de Secuencia Molecular
15.
Artículo en Inglés | MEDLINE | ID: mdl-28533887

RESUMEN

BACKGROUND: Malignant glioma is the most common brain cancer with devastating prognosis. Recurrence of malignant glioma following surgery is very common with few preventive and therapeutic options. Novel targets and therapeutic agents are constantly sought for better outcome. Our previous study established that inhibition of transient receptor potential melastatin 7 (TRPM7) channels resulted in significant decrease of human glioma cell growth and proliferation. As local anesthetic lidocaine has been shown to inhibit TRPM7 currents, we hypothesize that lidocaine may suppress glioma cell proliferation through TRPM7 channel inhibition. METHODS: TRPM7 currents were recorded in rat C6 glioma cells using the whole cell patch clamp technique. Cell growth and proliferation were assessed under microscopic examination and biochemical assays. RESULTS: Lidocaine inhibits TRPM7-like currents in a dose-dependent and reversible manner. At 1 and 3 mM, it inhibits ~30% and ~50% of TRPM7 currents. At these concentrations, it is effective in inhibiting the proliferation of C6 cells. As expected, the TRPM7 inhibitors gadolinium and 2-Aminoethoxydiphenyl borate have similar effects on TRPM7 currents and proliferation of C6 cells. Similar to its effect on C6 cells, lidocaine inhibits the proliferation of A172 cells, a human glioblastoma cell line. CONCLUSIONS: Lidocaine significantly inhibits the proliferation of glioma cells. The effect of lidocaine is mediated, at least in part, by inhibiting TRPM7 channels.

16.
Int J Pharm ; 513(1-2): 347-356, 2016 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-27628782

RESUMEN

Hydroxypropyl-ß-cyclodextrin (HP-ß-CD) has been widely used as an effective solubilizing agent in pharmaceutical industry for many years. However, the effect of degree of substitution (D.S.) of HP-ß-CD on solubilizing capacity and toxicity has not been concerned. In this study, solubilizing capacity of HP-ß-CDs with three different D.S. (4.55, 6.16 and 7.76) for 16 drugs were measured and their toxicities were compared by a 7-day i.v. administration (q.d.) study in rats. Generally, HP-ß-CD with high D.S. (7.76) showed weaker solubilizing capacity for steroids and BCS class II drugs, but lower hemolytic activity, compared with that of HP-ß-CD with low (4.55) or medium (6.16) D.S. HP-ß-CD with low D.S. resulted in more changes in hematological and biochemical parameters, but the effects were reversible after a 7-day recovery. Moreover, HP-ß-CD with medium D.S. may have slightly greater nephrotoxicity than the other two HP-ß-CDs. HP-ß-CDs with different D.S. had similar urine excretion percentage after i.v. administration and none of them was found to affect glomerular filtration function of rats. The results suggest that HP-ß-CD with low D.S. would be a better choice considering both the solubilizing capacity and toxicity. However, comparison in toxicity of HP-ß-CDs with different D.S. should be carried out in human in view of its species-dependence property.


Asunto(s)
Excipientes/química , Excipientes/toxicidad , beta-Ciclodextrinas/química , beta-Ciclodextrinas/toxicidad , 2-Hidroxipropil-beta-Ciclodextrina , Animales , Eritrocitos/efectos de los fármacos , Excipientes/farmacocinética , Femenino , Tasa de Filtración Glomerular/efectos de los fármacos , Hemólisis/efectos de los fármacos , Masculino , Preparaciones Farmacéuticas/química , Conejos , Ratas Sprague-Dawley , Solubilidad , beta-Ciclodextrinas/farmacocinética , beta-Ciclodextrinas/orina
17.
Drug Des Devel Ther ; 10: 3789-3798, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27895469

RESUMEN

Androst-3ß,5α,6ß-triol (Triol) is a promising neuroprotective agent, but its poor solubility restricts its development into parenteral preparations. In this study, Triol is significantly solubilized by bile salt/phosphatidylcholine mixed micelles (BS/PC-MM). All BS/PC-MM systems are tested to remarkably improve the drug solubility with various stabilities after drug loading. Among them, the sodium glycocholate (SGC)/egg phosphatidylcholine (EPC) system with 2:1 ratio in weight and the total concentration of SGC and EPC of 100 mg/mL is proved to produce stable mixed micelles with high drug loading. It is found that the stability of drug-loaded mixed micelles is quite different, which might be related to the change in critical micelle concentration (CMC) after incorporating drugs. SGC/EPC and SGC/soya phosphatidylcholine (SPC) remain transparent under accelerated conditions and manifest a decreased CMC (dropping from 0.105 to 0.056 mg/mL and from 0.067 to 0.024 mg/mL, respectively). In contrast, swine bile acid-sodium salt (SBA-Na)/PC and sodium deoxycholate (SDC)/PC are accompanied by drug precipitation and reached the maximum CMC on the first and the third days, respectively. Interestingly, the variation of CMC under accelerated testing conditions highly matches the drug-precipitating event in the primary stability experiment. In brief, the bile salt/phosphatidylcholine system exists as a potential strategy of improving sterol drug solubility. CMC variation under accelerated testing conditions might be a simple and easy method to predict the stability of drug-loaded mixed micelles.


Asunto(s)
Ácidos y Sales Biliares/química , Fosfatidilcolinas/química , Fosfatidilcolinas/farmacología , Esteroles/química , Esteroles/farmacología , Animales , Estabilidad de Medicamentos , Micelas , Solubilidad , Porcinos
18.
Mol Med Rep ; 13(3): 2499-505, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26821268

RESUMEN

Glutamate is the principal neurotransmitter in the central nervous system. Glutamate-mediated excitotoxicity is the predominant cause of cerebral damage. Recent studies have shown that lysosomal membrane permeabilization (LMP) is involved in ischemia­associated neuronal death in non­human primates. This study was designed to investigate the effect of glutamate on lysosomal stability in primary cultured cortical neurons. Glutamate treatment for 30 min induced the permeabilization of lysosomal membranes as assessed by acridine orange redistribution and immunofluorescence of cathepsin B in the cytoplasm. Inhibition of glutamate excitotoxicity by the NMDA receptor antagonist MK­801 and the calcium chelator ethylene glycol­bis (2­aminoethylether)­N, N, N', N'­tetraacetic acid, rescued lysosomes from permeabilization. The role of calpain and reactive oxygen species (ROS) in inducing LMP was also investigated. Ca2+ overload following glutamate treatment induced the activation of calpain and the production of ROS, which are two major contributors to neuronal death. It has been reported that lysosomal­associated membrane protein 2 (LAMP2) and heat shock protein (HSP)70 are two calpain substrates that promote LMP in cancer cells; however, it was found that calpains were activated by glutamate, but only LAMP2 was subsequently degraded. Furthermore, LMP was not alleviated by treatment with the calpain inhibitors calpeptin and SJA6017, which blocked the cleavage of the calpain substrate α­fodrin. It was demonstrated that LMP was significantly alleviated by treatment with the antioxidant N­Acetyl­L­cysteine, indicating that LMP involvement in early glutamate excitotoxicity may be mediated partly by ROS rather than calpain activation. Overall, these data shed light on the role of ROS-mediated LMP in early glutamate excitotoxicity.


Asunto(s)
Ácido Glutámico/farmacología , Lisosomas/metabolismo , Neuronas/metabolismo , Animales , Calpaína/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Lisosomas/efectos de los fármacos , Cultivo Primario de Células , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo
19.
Neuroreport ; 16(13): 1461-5, 2005 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-16110272

RESUMEN

In this study, we have investigated the role of albumin in zinc-induced neurotoxicity of cultured rat cerebellar granule neurons. ZnCl2 induces cerebellar granule cell death in a time- and concentration-dependent manner and albumin in serum affords a significant protection against zinc-induced neurotoxicity. We further measured intracellular zinc concentrations in the presence or absence of albumin. The results showed that exogenously added ZnCl2 induced an increase in intracellular zinc concentrations in a concentration-dependent fashion in albumin-free buffer. However, in the presence of albumin, it dramatically decreased ZnCl2-induced elevation of intracellular zinc concentrations. These results indicate that albumin protects against zinc-induced neurotoxicity by blocking extracellular zinc uptake by neurons.


Asunto(s)
Albúminas/farmacología , Cloruros/toxicidad , Neuronas/citología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Compuestos de Zinc/toxicidad , Animales , Células Cultivadas , Cerebelo/citología , Cloruros/farmacocinética , Medios de Cultivo/farmacología , Interacciones Farmacológicas , Ratas , Ratas Sprague-Dawley , Compuestos de Zinc/farmacocinética
20.
Neurosci Lett ; 353(3): 209-12, 2003 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-14665418

RESUMEN

Brain damage induced by unconjugated bilirubin, the end product of heme catabolism, in human neonates is a well recognized clinical syndrome. However, the cellular and molecular mechanisms underlying bilirubin neurotoxicity remain unclear. To characterize the sequence of events leading to bilirubin-induced neurotoxicity, we have exposed rat cerebellar granule neurons (CGN) to bilirubin and investigated whether activation of p38 MAP kinase mediates neuronal death. In this study, bilirubin markedly induces an early activation of p38 MAP kinase at 1 h. Pretreatment of neurons with a p38 MAP kinase inhibitor, SB 203580, significantly protected CGN against bilirubin-induced neurotoxicity. Our data suggest that hyperphosphorylation of p38 MAP kinase plays an important role in bilirubin-induced neuronal death and provides a novel approach to discovering drugs to treat bilirubin-induced encephalopathy.


Asunto(s)
Bilirrubina/farmacología , Muerte Celular/efectos de los fármacos , Cerebelo/citología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neuronas/efectos de los fármacos , Análisis de Varianza , Animales , Animales Recién Nacidos , Antracenos/farmacología , Western Blotting , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Imidazoles/farmacología , Neuronas/metabolismo , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA