Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Phytomedicine ; 112: 154700, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36774842

RESUMEN

BACKGROUND: Diabetic kidney disease (DKD) is a primary microvascular complication of diabetes. However, a complete cure for DKD has not yet been found. Although there is evidence that Rutin can delay the onset of DKD, the underlying mechanism remains unclear. PURPOSE: To investigate the renoprotective effect of Rutin in the process of DKD and to explore its potential molecular mechanisms. METHODS: Db/db mice and high glucose (HG)-induced human renal glomerular endothelial cells (GEnCs) were used as in vivo and in vitro models, respectively. Western blot (WB), Immunohistochemistry (IHC)and Immunofluorescence (IF) staining were used to identify the expression level of proteins associated with endothelial-to-mesenchymal transition (EndMT) and autophagy. Tandem Mass Tag (TMT)-based proteomics analysis was utilized to reveal the mechanism of Rutin in DKD. Transfection with small interfering RNA (siRNA) to reveal the role of histone deacetylase 1 (HDAC1) in HG-induced GEnCs. RESULTS: Following 8 weeks of Rutin administration, db/db mice's kidney function and structure significantly improved. In HG-induced GEnCs, activation of autophagy attenuates cellular EndMT. Rutin could alleviate EndMT and restore autophagy in vivo and in vitro models. Proteomics analysis results showed that HDAC1 significantly downregulated in the 200 mg/kg/d Rutin group compared with the db/db group. Transfection with si-HDAC1 in GEnCs partially blocked HG-induced EndMT and restored autophagy. Furthermore, Rutin inhibits the phosphorylation of the PI3K / AKT/ mTOR pathway. HDAC1 overexpression was suppressed in HG-induced GEnCs after using Rapamycin, a specific mTOR inhibitor, verifying the correlation between mTOR and HDAC1. CONCLUSION: Rutin alleviates EndMT by restoring autophagy through inhibiting HDAC1 via the PI3K/AKT/mTOR pathway in DKD.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Ratones , Animales , Humanos , Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Células Endoteliales/metabolismo , Histona Desacetilasa 1/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Autofagia
2.
Toxicology ; 478: 153293, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35995123

RESUMEN

Cassiae semen (CS), a traditional Chinese medicine, has various bioactivities in preclinical and clinical practice. Aurantio-obtusin (AO) is a major anthraquinone (AQ) ingredient derived from CS, and has drawn public concerns over its potential hepatotoxicity. We previously found that AO induces hepatic necroinflammation by activating NOD-like receptor protein 3 inflammasome signaling. However, the mechanisms contributing to AO-motivated hepatotoxicity remain unclear. Herein, we evaluated hepatotoxic effects of AO on three liver cell lines by molecular and biochemical analyses. We found that AO caused cell viability inhibition and biochemistry dysfunction in the liver cells. Furthermore, AO elevated reactive oxygen species (ROS), followed by mitochondrial dysfunction (decreases in mitochondrial membrane potential and adenosine triphosphate) and apoptosis (increased Caspase-3, Cleaved caspase-3, Cytochrome c and Bax expression, and decreased Bcl-2 expression). We also found that AO increased the lipid peroxidation (LPO) and enhanced ferroptosis by activating cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA)-cAMP response element-binding (CREB) pathway (increases in PKA, p-CREB, acyl-CoA synthetase long chain family member 4). Based on these results, we used an AOP framework to explore the mechanisms underlying AO's hepatotoxicity. It starts from molecular initiating event (ROS), and follows two critical toxicity pathways (i.e., mitochondrial dysfunction-mediated apoptosis and LPO-enhanced ferroptosis) over a series of key events (KEs) to the adverse outcome of hepatotoxicity. The results of an assessment confidence in the adverse outcome pathway (AOP) framework supported the evidence concordance in dose-response, temporal and incidence relationships between KEs in AO-induced hepatotoxicity. This study's findings offer a novel toxicity pathway network for AO-caused hepatotoxicity.


Asunto(s)
Rutas de Resultados Adversos , Enfermedad Hepática Inducida por Sustancias y Drogas , Antraquinonas/química , Antraquinonas/farmacología , Caspasa 3 , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Humanos , Especies Reactivas de Oxígeno
3.
Front Cell Dev Biol ; 9: 632843, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33791294

RESUMEN

Alzheimer's disease (AD), one of the most common neurodegenerative diseases, has no effective treatment. We studied the potential effects of tetramethylpyrazine (TMP), an alkaloid in the rhizome of Ligusticum chuanxiong Hort. used in Traditional Chinese Medicine (chuanxiong) to treat ischemic stroke, on AD progression in two AD mouse models. Eight-month-old 3xTg-AD mice received TMP treatment (10 mg/kg/d) for 1 month, and 4-month-old APP/PS1-AD mice received TMP treatment (10 mg/kg/d) for 2 months. Behavioral tests, including step-down passive avoidance (SDA), new object recognition (NOR), Morris water maze (MWM), and Contextual fear conditioning test showed that TMP significantly improved the learning and memory of the two AD-transgenic mice. In addition, TMP reduced beta-amyloid (Aß) levels and tau phosphorylation (p-tau). Venny map pointed out that 116 proteins were commonly changed in 3xTg mice vs. wild type (WT) mice and TMP-treated mice vs. -untreated mice. The same 130 proteins were commonly changed in APP/PS1 mice vs. WT mice and TMP-treated mice vs. -untreated mice. The functions of the common proteins modified by TMP in the two models were mainly involved in mitochondrial, synaptic, cytoskeleton, ATP binding, and GTP binding. Mitochondrial omics analysis revealed 21 and 20 differentially expressed mitochondrial proteins modified by TMP in 3xTg-AD mice and APP/PS1 mice, respectively. These differential proteins were located in the mitochondrial inner membrane, mitochondrial outer membrane, mitochondrial gap, and mitochondrial matrix, and the function of some proteins is closely related to oxidative phosphorylation (OXPHOS). Western-blot analysis confirmed that TMP changed the expression of OXPHOS complex proteins (sdhb, ndufa10, uqcrfs1, cox5b, atp5a) in the hippocampus of the two AD mice. Taken together, we demonstrated that TMP treatment changed the hippocampal proteome, reduced AD pathology, and reduced cognitive impairment in the two AD models. The changes might be associated with modification of the mitochondrial protein profile by TMP. The results of the study suggest that TMP can improve the symptoms of AD.

4.
Autophagy ; 17(11): 3833-3847, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33622188

RESUMEN

Alzheimer disease (AD) is the most prevalent neurodegenerative disorder leading to dementia in the elderly. Unfortunately, no cure for AD is available to date. Increasing evidence has proved the roles of misfolded protein aggregation due to impairment of the macroautophagy/autophagy-lysosomal pathway (ALP) in the pathogenesis of AD, and thus making TFEB (transcription factor EB), which orchestrates ALP, as a promising target for treating AD. As a complementary therapy, acupuncture or electroacupuncture (EA) has been commonly used for treating human diseases. Although the beneficial effects of acupuncture for AD have been primarily studied both pre-clinically and clinically, the real efficacy of acupuncture on AD remains inconclusive and the underlying mechanisms are largely unexplored. In this study, we demonstrated the cognitive-enhancing effect of three-needle EA (TNEA) in an animal model of AD with beta-amyloid (Aß) pathology (5xFAD). TNEA reduced APP (amyloid beta (A4) precursor protein), C-terminal fragments (CTFs) of APP and Aß load, and inhibited glial cell activation in the prefrontal cortex and hippocampus of 5xFAD. Mechanistically, TNEA activated TFEB via inhibiting the AKT-MAPK1-MTORC1 pathway, thus promoting ALP in the brains. Therefore, TNEA represents a promising acupuncture therapy for AD, via a novel mechanism involving TFEB activation.Abbreviations Aß: ß-amyloid; AD: Alzheimer disease; AIF1/IBA1: allograft inflammatory factor 1; AKT1: thymoma viral proto-oncogene 1; ALP: autophagy-lysosomal pathway; APP: amyloid beta (A4) precursor protein; BACE1: beta-site APP cleaving enzyme 1; CQ: chloroquine; CTFs: C-terminal fragments; CTSD: cathepsin D; EA: electroacupuncture; FC: fear conditioning; GFAP: glial fibrillary acidic protein; HI: hippocampus; LAMP1: lysosomal-associated membrane protein 1; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; MAPK1/ERK2: mitogen-activated protein kinase 1; MAPT: microtubule-associated protein tau; MTORC1: mechanistic target of rapamycin kinase complex 1; MWM: Morris water maze; NFT: neurofibrillary tangles; PFC: prefrontal cortex; PSEN1: presenilin 1; SQSTM1/p62: sequestosome 1; TFEB: transcription factor EB; TNEA: three-needle electroacupuncture.


Asunto(s)
Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/metabolismo , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Encéfalo/patología , Disfunción Cognitiva/terapia , Electroacupuntura , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Encéfalo/metabolismo , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , Modelos Animales de Enfermedad , Electroacupuntura/métodos , Femenino , Lisosomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Microglía/patología , Prueba del Laberinto Acuático de Morris
5.
J Mol Neurosci ; 71(7): 1456-1466, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33403592

RESUMEN

T-006, a small-molecule compound derived from tetramethylpyrazine (TMP), has potential for the treatment of neurological diseases. In order to investigate the effect of T-006 prophylactic treatment on an Alzheimer's disease (AD) model and identify the target of T-006, we intragastrically administered T-006 (3 mg/kg) to Alzheimer's disease (AD) transgenic mice (APP/PS1-2xTg and APP/PS1/Tau-3xTg) for 6 and 8 months, respectively. T-006 improved cognitive ability after long-term administration in two AD mouse models and targeted mitochondrial-related protein alpha-F1-ATP synthase (ATP5A). T-006 significantly reduced the expression of phosphorylated-tau, total tau, and APP while increasing the expression of synapse-associated proteins in 3xTg mice. In addition, T-006 modulated the JNK and mTOR-ULK1 pathways to reduce both p-tau and total tau levels. Our data suggested that T-006 mitigated cognitive decline primarily by reducing the p-tau and total tau levels in 3xTg mice, supporting further investigation into its development as a candidate drug for AD treatment.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Trastornos del Conocimiento/tratamiento farmacológico , Hidrazonas/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Pirazinas/uso terapéutico , Proteínas tau/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Autofagia/efectos de los fármacos , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Reacción de Prevención , Modelos Animales de Enfermedad , Donepezilo/farmacología , Donepezilo/uso terapéutico , Evaluación Preclínica de Medicamentos , Hidrazonas/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Memantina/farmacología , Memantina/uso terapéutico , Ratones , Ratones Transgénicos , Prueba del Laberinto Acuático de Morris , Fármacos Neuroprotectores/farmacología , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Pirazinas/farmacología , Distribución Aleatoria , Reconocimiento en Psicología , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
6.
Nutr Neurosci ; 24(9): 720-734, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31603034

RESUMEN

Effective treatment to prevent or arrest the advance of Alzheimer disease (AD) has yet to be discovered. We investigated whether OligonolR, an FDA-approved flavanol-rich extract prepared from lychee fruit and green tea, exerted beneficial effects relevant to AD in a triple transgenic male mouse model of AD (3×Tg-AD). At 9 months of age, untreated 3×Tg-AD mice vs. wild-type (WT) controls displayed cognitive deficits in behavioral assays and, at 12 months, elevated levels of hippocampal amyloid beta-protein (Aß), amyloid precursor protein (APP), tau phosphorylation, and pro-inflammatory cytokines. 3×Tg-AD mice given Oligonol showed fewer cognitive deficits and attenuated pathological indices at 12 months. Oligonol treatment of 3×Tg-AD mice modulated expression of some critical brain proteins that involve multiple pathways relevant to mitochondrial dysfunction, proteasomal failure, endoplasmic reticulum (ER) stress and synaptic impairment. Together, these results demonstrate that continuous Oligonol treatment attenuates AD-like pathology and cognitive impairment of 3×Tg-AD mice and set the stage for clinical trials of this flavanol-rich plant extract in patients with early AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Catequina/análogos & derivados , Disfunción Cognitiva/tratamiento farmacológico , Litchi/química , Fenoles/administración & dosificación , Extractos Vegetales/administración & dosificación , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Catequina/administración & dosificación , Modelos Animales de Enfermedad , Flavonoides/administración & dosificación , Frutas/química , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Masculino , Ratones , Ratones Transgénicos , Mutación/genética , Fosforilación/efectos de los fármacos , Té/química , Transgenes/genética , Proteínas tau/genética , Proteínas tau/metabolismo
7.
J Diabetes Res ; 2020: 9309768, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32051833

RESUMEN

Diabetic kidney disease (DKD) is a major cause of end-stage renal disease (ESRD), and therapeutic strategies for delaying its progression are limited. Loss of podocytes by apoptosis characterizes the early stages of DKD. To identify novel therapeutic options, we investigated the effects of Xuesaitong (XST), consisting of total saponins from Panax notoginseng, on podocyte apoptosis in streptozotocin- (STZ-) induced diabetic rats. XST (5 mg/kg·d) or Losartan (10 mg/kg·d) was given to diabetic rats for 12 weeks. Albuminuria, renal function markers, and renal histopathology morphological changes were examined. Podocyte apoptosis was determined by triple immunofluorescence labelling including a TUNEL assay, WT1, and DAPI. Renal expression of Nox4, miRNA-214, PTEN, PDK1, phosphorylated Akt, mTOR, and mTORC1 was detected. In diabetic rats, severe hyperglycaemia and albuminuria developed, and apoptotic podocytes were markedly increased in diabetic kidneys. However, XST attenuated albuminuria, mesangial expansion, podocyte apoptosis, and morphological changes of podocytes in diabetic rats. Decreased expression of PTEN, as well as increased expression of Nox4, miRNA-214, PDK1, phosphorylated Akt, mTOR, and mTORC1, was detected. These abnormalities were partially restored by XST treatment. Thus, XST ameliorated podocyte apoptosis partly through modulating the PTEN-PDK1-Akt-mTOR pathway. These novel findings might point the way to a natural therapeutic strategy for treating DKD.


Asunto(s)
Apoptosis/efectos de los fármacos , Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/metabolismo , Medicamentos Herbarios Chinos/farmacología , Podocitos/efectos de los fármacos , Sustancias Protectoras/farmacología , Saponinas/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Fosfohidrolasa PTEN/metabolismo , Podocitos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/metabolismo , Ratas , Serina-Treonina Quinasas TOR/metabolismo
8.
Br J Pharmacol ; 176(17): 3318-3335, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31180578

RESUMEN

BACKGROUND AND PURPOSE: Cerebral vasospasm and neuronal apoptosis after subarachnoid haemorrhage (SAH) is the major cause of morbidity and mortality in SAH patients. So far, single-target agents have not prevented its occurrence. Memantine, a non-competitive NMDA re3ceptor antagonist, is known to alleviate brain injury and vasospasm in experimental models of SAH. Impairment of NO availability also contributes to vasospasm. Recently, we designed and synthesized a memantine nitrate MN-08, which has potent dual functions: neuroprotection and vasodilation. Here, we have tested the therapeutic effects of MN-08 in animal models of SAH. EXPERIMENTAL APPROACH: Binding to NMDA receptors (expressed in HEK293 cells), NO release and vasodilator effects of MN-08 were assessed in vitro. Therapeutic effects of MN-08 were investigated in vivo, using rat and rabbit SAH models. KEY RESULTS: MN-08 bound to the NMDA receptor, slowly releasing NO in vitro and in vivo. Consequently, MN-08 relaxed the pre-contracted middle cerebral artery ex vivo and increased blood flow velocity in small vessels of the mouse cerebral cortex. It did not, however, lower systemic blood pressure. In an endovascular perforation rat model of SAH, MN-08 improved the neurological scores and ameliorated cerebral vasospasm. Moreover, MN-08 also alleviated cerebral vasospasm in a cisterna magna single-injection model in rabbits. MN-08 attenuated neural cell apoptosis in both rat and rabbit models of SAH. Importantly, the therapeutic benefit of MN-08 was greater than that of memantine. CONCLUSION AND IMPLICATIONS: MN-08 has neuroprotective potential and can ameliorate vasospasm in experimental SAH models.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Modelos Animales de Enfermedad , Memantina/uso terapéutico , Nitratos/uso terapéutico , Hemorragia Subaracnoidea/tratamiento farmacológico , Vasodilatadores/uso terapéutico , Vasoespasmo Intracraneal/tratamiento farmacológico , Animales , Lesiones Encefálicas/inducido químicamente , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Masculino , Memantina/administración & dosificación , Memantina/química , Ratones , Ratones Endogámicos C57BL , Nimodipina , Nitratos/administración & dosificación , Nitratos/química , Óxido Nítrico/análisis , Conejos , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad , Hemorragia Subaracnoidea/inducido químicamente , Vasodilatadores/administración & dosificación , Vasodilatadores/química , Vasoespasmo Intracraneal/inducido químicamente
9.
Circulation ; 139(19): 2260-2277, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-30773021

RESUMEN

BACKGROUND: Elevated levels of S-adenosylhomocysteine (SAH), the precursor of homocysteine, are positively associated with the risk of cardiovascular disease and with the development and progression of atherosclerosis. However, the role of SAH in endothelial dysfunction is unclear. METHODS: Apolipoprotein E-deficient ( apoE-/-) mice received dietary supplementation with the SAH hydrolase (SAHH) inhibitor adenosine dialdehyde or were intravenously injected with a retrovirus expressing SAHH shRNA. These 2 approaches, along with the heterozygous SAHH gene knockout ( SAHH+/-) mouse model, were used to elevate plasma SAH levels and to examine the role of SAH in aortic endothelial dysfunction. The relationship between plasma SAH levels and endothelial dysfunction was also investigated in human patients with coronary artery disease and healthy control subjects. RESULTS: Plasma SAH levels were increased in SAHH+/- mice and in apoE-/- mice after dietary administration of adenosine dialdehyde or intravenous injection with SAHH shRNA. SAHH+/- mice or apoE-/- mice with SAHH inhibition showed impaired endothelium-dependent vascular relaxation and decreased nitric oxide bioavailability after treatment with acetylcholine; this was completely abolished by the administration of the endothelial nitric oxide synthase inhibitor NG-nitro-l-arginine methyl ester. Furthermore, SAHH inhibition induced production of reactive oxygen species and p66shc expression in the mouse aorta and human aortic endothelial cells. Antioxidants and p66shc siRNA prevented SAHH inhibition-induced generation of reactive oxygen species and attenuated the impaired endothelial vasomotor responses in high-SAH mice. Moreover, inhibition of SAHH induced hypomethylation in the p66shc gene promoter and inhibited expression of DNA methyltransferase 1. Overexpression of DNA methyltransferase 1, induced by transduction of an adenovirus, was sufficient to abrogate SAHH inhibition-induced upregulation of p66shc expression. Finally, plasma SAH levels were inversely associated with flow-mediated dilation and hypomethylation of the p66shc gene promoter and positively associated with oxidative stress levels in patients with coronary artery disease and healthy control subjects. CONCLUSIONS: Our findings indicate that inhibition of SAHH results in elevated plasma SAH levels and induces endothelial dysfunction via epigenetic upregulation of the p66shc-mediated oxidative stress pathway. Our study provides novel molecular insight into mechanisms of SAH-associated endothelial injury that may contribute to the development of atherosclerosis. CLINICAL TRIAL REGISTRATION: URL: https://www.clinicaltrials.gov . Unique identifier: NCT03345927.


Asunto(s)
Adenosilhomocisteinasa/metabolismo , Aterosclerosis/metabolismo , Enfermedad de la Arteria Coronaria/metabolismo , Endotelio Vascular/fisiología , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo , Adenosina/administración & dosificación , Adenosina/análogos & derivados , Adenosina/farmacología , Adenosilhomocisteinasa/antagonistas & inhibidores , Adenosilhomocisteinasa/genética , Anciano , Animales , Metilación de ADN , Modelos Animales de Enfermedad , Epigénesis Genética , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Persona de Mediana Edad , Estrés Oxidativo , ARN Interferente Pequeño/genética , S-Adenosilhomocisteína/sangre , Transducción de Señal , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/genética
10.
RSC Adv ; 9(33): 18776-18784, 2019 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-35516848

RESUMEN

Alzheimer's disease (AD) is a common neurodegenerative disease characterized by pathological processes, including abnormal amyloid deposits and filament tangles, oxidative stress, neuroinflammation, and neurotrophic insufficiency, leading to chronic and prolonged neuronal loss and cognitive deficits. Tetramethylpyrazine (TMP) is one of the main active components of Ligusticum wallichii, a traditional Chinese medicine widely used for brain related disease. Here, we synthesized the TMP derivative tetramethylpyrazine dimer (DTMP), and evaluated the potential mechanisms underlying its potential neuroprotective effects using the murine neuron-like cells (N2a) transfected with the human "Swedish" mutant amyloid precursor protein (N2aAPP). ELISA results indicated that DTMP reduced the levels of Aß1-40 and Aß1-42 in N2aAPP. Then through proteomic analysis we identified a total of 208 differentially expressed proteins in N2aAPP cells compared to the wild-type N2a cells (N2aWT), including 144 increased and 64 decreased proteins. 449 differentially expressed proteins were revealed in N2aAPP cells on DTMP treatment with 69 increased and 380 decreased proteins. Bioinformatic analysis suggested that these proteins are enriched in mitochondrial function, the electronic transmission chain, ATP binding, oxidative phosphorylation, GTPase function, the transcriptional translation process, amino acid metabolism, nucleotide binding and others. Given the vital role of mitochondria in the pathogenesis of AD, we selected the electron transport chain pathway-related molecules to further validate these findings. Western-blot analysis demonstrated that DTMP significantly increased the levels of complex I (NDUAA), complex II (SDHB), complex III (UCRI), complex IV (COX5A) and complex V (ATP5A) in N2aAPP cells. The modulation of dysregulated proteins implicated in AD pathogenesis implies the pharmacological mechanisms of DTMP and its potential as a novel therapeutic choice in AD.

11.
Neuromolecular Med ; 20(1): 97-111, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29411248

RESUMEN

Our previous studies demonstrated that the multifunctional agent TBN, a derivative of tetramethylpyrazine armed with a nitrone moiety, displayed high therapeutic efficacy in experimental ischemic stroke models. However, its molecular mechanisms of action underlying the neuroprotective effect need further exploration. In the present study, we found that TBN had significant activities scavenging free radicals such as ·OH, O 2·- and ONOO-, inhibiting Ca2+ overload, maintaining mitochondrial function and preventing neuronal damage in primary cortical cultures. Further, TBN was effective in reducing brain infarction and ameliorating impairment of behavioral functions in the permanent middle cerebral artery occlusion (p-MCAo) rat model. TBN down-regulated the expression of pro-apoptotic factors Bax, while up-regulated the expression of anti-apoptotic factor Bcl-2 and increased the expression of pro-survival factors including p-Akt and p-GSK3ß in the peri-infarct cortex of p-MCAo rats. In addition, LY-294002 (a PI3K inhibitor) and MK2206 (an Akt inhibitor) significantly blocked the protective effect of TBN against OGD-induced death of cortical neurons. Taken together, the multifunctional mechanisms including scavenging free radicals, blocking calcium overload, maintaining mitochondrial function and activating the PI3K/Akt/p-GSK3ß cell survival pathway were possibly involved in the neuroprotective effects of TBN, making it a promising clinical candidate for the treatment of ischemic stroke.


Asunto(s)
Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Óxidos de Nitrógeno/uso terapéutico , Pirazinas/uso terapéutico , Animales , Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteínas Reguladoras de la Apoptosis/genética , Calcio/metabolismo , Hipoxia de la Célula , Células Cultivadas , Evaluación Preclínica de Medicamentos , Depuradores de Radicales Libres/farmacología , Depuradores de Radicales Libres/uso terapéutico , Glucosa/farmacología , Ácido Glutámico/toxicidad , Infarto de la Arteria Cerebral Media/prevención & control , Peroxidación de Lípido/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Actividad Motora/efectos de los fármacos , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Fármacos Neuroprotectores/farmacología , Óxidos de Nitrógeno/farmacología , Técnicas de Placa-Clamp , Inhibidores de Proteínas Quinasas/farmacología , Pirazinas/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
12.
Toxicol Res (Camb) ; 5(4): 1130-1139, 2016 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-30090419

RESUMEN

Recent studies suggest that copper exposure, even at very low levels, can produce significant toxic effects on the brains of mice. This study is aimed to explore the effects of low levels of copper on the hippocampal proteome of mice. Two-dimensional fluorescence difference gel electrophoresis was performed on hippocampal homogenate obtained from mice, which were given either drinking water only (control) or water supplemented with 0.13 ppm copper (copper-treated) for a period of 8 months beginning at an age of 3 months. A total of 9 differentially expressed proteins between copper-treated mice and control mice were identified. Protein functional analysis revealed that the altered proteins mainly involved energy metabolism-related proteins, synaptic proteins, molecular chaperones and cellular structural components. Among these differentially expressed proteins, serine racemase (SRR) and glial fibrillary acidic protein (GFAP) were significantly down-regulated and up-regulated, respectively, in the hippocampus of copper-treated mice compared with the control mice. SRR was shown to be involved in memory formation. The increased expression of GFAP, an astrocyte marker, indicated that long-term low levels of copper exposure caused activation of the inflammatory response, a process linked to spatial memory impairment. In agreement with the data from proteomic analysis, memory impairment was observed in copper-treated mice as measured by the Morris water maze test. In summary, this study has identified a number of abnormally expressed proteins in the hippocampus of copper-treated mice, and the identified protein, such as SRR, together with inflammatory responses, as evidenced by the increased expression of GFAP, could contribute to memory impairment resulting from copper exposure. Our findings provide insights for a better understanding of copper neurotoxicity at the protein level in response to low levels of copper exposure.

13.
Pflugers Arch ; 462(4): 587-97, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21796340

RESUMEN

This article presents a novel model of acupuncture physiology based on cellular calcium activation by an acoustic shear wave (ASW) generated by the mechanical movement of the needle. An acupuncture needle was driven by a piezoelectric transducer at 100 Hz or below, and the ASW in human calf was imaged by magnetic resonance elastography. At the cell level, the ASW activated intracellular Ca(2+) transients and oscillations in fibroblasts and endothelial, ventricular myocytes and neuronal PC-12 cells along with frequency-amplitude tuning and memory capabilities. Monitoring in vivo mammalian experiments with ASW, enhancement of endorphin in blood plasma and blocking by Gd(3+) were observed; and increased Ca(2+) fluorescence in mouse hind leg muscle was imaged by two-photon microscopy. In contrast with traditional acupuncture models, the signal source is derived from the total acoustic energy. ASW signaling makes use of the anisotropy of elasticity of tissues as its waveguides for transmission and that cell activation is not based on the nervous system.


Asunto(s)
Estimulación Acústica , Terapia por Acupuntura , Señalización del Calcio/fisiología , Músculo Esquelético/fisiología , Adulto , Animales , Anisotropía , Diagnóstico por Imagen de Elasticidad , Humanos , Masculino , Ratones , Modelos Teóricos , Células 3T3 NIH , Células PC12 , Ratas , Muslo
14.
J Alzheimers Dis ; 19(3): 813-27, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20157238

RESUMEN

Previous clinical and epidemiological studies have suggested that elevated plasma homocysteine (Hcy) levels increased the risk of Alzheime's disease (AD). Although the underlying mechanisms of its toxicity are elusive, it has been shown that Hcy damages neurons by inducing apoptosis, DNA fragmentation, and tau hyperphosphorylation. Wolfberry (Lycium barbarum) is a fruit that is known for its eye-protective and anti-aging properties in Asian countries. Previous studies from our laboratory have demonstrated that polysaccharides derived from wolfberry (LBA) have the ability to protect neurons from amyloid-beta (Abeta) peptide neurotoxicity. We hypothesize that the neuroprotective effects of wolfberry is not limited to Abeta and can also provide protection against other AD risk factors. In this study, we aim to elucidate the neuroprotective effects of wolfberry against Hcy-induced neuronal damage. Our data showed that LBA treatment significantly attenuated Hcy-induced neuronal cell death and apoptosis in primary cortical neurons as demonstrated by LDH and caspase-3 like activity assay. LBA also significantly reduced Hcy-induced tau phosphorylation at tau-1 (Ser198/199/202), pS396 (Ser396), and pS214 (Ser214) epitopes as well as cleavage of tau. At the same time, we also found that the phosphorylation level of p-GSK3beta (Ser9/Tyr 216) remained unchanged among different treatment groups at all detected time points. LBA treatment suppressed elevation of both p-ERK and p-JNK. In summary, our data demonstrated that LBA exerted neuroprotective effects on cortical neurons exposed to Hcy. Therefore, LBA has the potential to be a diseasemodifying agent for the prevention of AD.


Asunto(s)
Enfermedad de Alzheimer/prevención & control , Corteza Cerebral , Medicamentos Herbarios Chinos/farmacología , Homocisteína/antagonistas & inhibidores , Homocisteína/toxicidad , Neuronas , Fármacos Neuroprotectores/farmacología , Fitoterapia , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Fragmentación del ADN , Medicamentos Herbarios Chinos/administración & dosificación , Hidroliasas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/administración & dosificación , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Proteínas tau/antagonistas & inhibidores
15.
Biofactors ; 32(1-4): 169-78, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19096113

RESUMEN

We tested the hypotheses that supplemental intake of the diet supplement Coenzyme Q10 (CoQ10) could delay brain atrophy in double transgenic amyloid precursor protein (APP) / presenilin 1 (PS1), single transgenic APP and PS1 as well as wild type mice by volume MR image in vivo. One hundred and twelve mice (28 APP/PS1, 28 APP, 28 PS1 and 28 wild types) were studied. Half of each genotype group (n = 14 per group) was treated with CoQ10 2400 mg/kg/day, and the other half with placebo for 60 days. Magnetic resonance (MR) images were used to obtain the volumes of the hemispheres and hippocampi. APP / PS1, APP, PS1 and wild type mice treated with CoQ10 exhibited significantly less atrophy in hemisphere and hippocampus than those receiving placebo. The neuro-protective effect of the CoQ10 on hemispheric volume, and hippocampal volume was related to genotype; greater in APP/PS1 than APP and PS1 mice and less in wild type mice. Our result indicated that CoQ10 may have therapeutic potential in the prevention and treatment of MCI and AD.


Asunto(s)
Envejecimiento/fisiología , Antioxidantes/uso terapéutico , Encéfalo/patología , Depuradores de Radicales Libres/uso terapéutico , Proteína Amiloide A Sérica/genética , Ubiquinona/análogos & derivados , Animales , Atrofia/prevención & control , Cerebro/anatomía & histología , Cerebro/efectos de los fármacos , Trastornos del Conocimiento/prevención & control , Femenino , Hipocampo/anatomía & histología , Hipocampo/efectos de los fármacos , Imagen por Resonancia Magnética/veterinaria , Masculino , Ratones , Ratones Transgénicos , Presenilina-1/genética , Ubiquinona/uso terapéutico
16.
J Mol Neurosci ; 34(2): 165-71, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18181031

RESUMEN

One of the neuropathological features of Alzheimer's disease (AD) is the deposition of senile plaques containing beta-amyloid (A beta). There is limited evidence for the treatment to arrest A beta pathology of AD. In our present study, we tested the effect of coenzyme Q10 (CoQ10), an endogenous antioxidant and a powerful free radical scavenger, on A beta in the aged transgenic mice overexpressing Alzheimer presenilin 1-L235P (leucine-to-proline mutation at codon 235, 16-17 months old). The treatment by feeding the transgenic mice with CoQ10 for 60 days (1,200 mg kg(-1) day(-1)) partially attenuated A beta overproduction and intracellular A beta deposit in the cortex of the transgenic mice compared with the age-matched untreated transgenic mice. Meanwhile, an increased oxidative stress reaction was detected as evidenced by elevated level of malondialdehyde (MDA) and decreased activity of superoxide dismutase (SOD) in the transgenic mice relative to the wild-type mice, and supplementation of CoQ10 partially decreased MDA level and upregulated the activity of SOD. The results indicate that oxidative stress is enhanced in the brain of the transgenic mice, that this enhancement may further promote A beta 42 overproduction in a vicious formation, and that CoQ10 would be beneficial for the therapy of AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Fragmentos de Péptidos/metabolismo , Presenilina-1/genética , Ubiquinona/análogos & derivados , Vitaminas/metabolismo , Anciano , Enfermedad de Alzheimer/metabolismo , Animales , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Coenzimas/metabolismo , Femenino , Humanos , Malondialdehído/metabolismo , Ratones , Ratones Transgénicos , Estrés Oxidativo , Placa Amiloide/patología , Mutación Puntual , Presenilina-1/metabolismo , Superóxido Dismutasa/metabolismo , Ubiquinona/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA