Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biomed Pharmacother ; 172: 116204, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38364733

RESUMEN

HACE1 is a member of the HECT domain-containing E3 ligases with 909 amino acid residues, containing N-terminal ankyrin-repeats (ANK) and C-terminal HECT domain. Previously, it was shown that HACE1 is inactive in human tumors and plays a crucial role in the initiation, progression, and invasion of malignant tumors. Recent studies indicated that HACE1 might be closely involved in neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease. HACE1 interacts with its substrates, including Ras-related C3 botulinum toxin substrate 1 (Rac1), nuclear factor erythroid 2-related factor 2 (Nrf2), tumor necrosis factor receptor (TNFR), and optineurin (OPTN), through which participates in several pathophysiological processes, such as oxidative stress, autophagy and inflammation. Therefore, in this review, we elaborately describe the essential substrates of HACE1 and illuminate the pathophysiological processes by which HACE1 is involved in neurodegenerative diseases. We provide a new molecular target for neurodegenerative diseases.


Asunto(s)
Enfermedades Neurodegenerativas , Ubiquitina-Proteína Ligasas , Humanos , Enfermedad de Alzheimer , Aminoácidos , Enfermedad de Huntington , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Enfermedad de Parkinson , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores
2.
J Asian Nat Prod Res ; 26(1): 146-153, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38419338

RESUMEN

White matter lesions (WMLs), characterized by focal demyelination or myelination disorders, are commonly present in cerebral small vessel disease and various neurological diseases. Multiple etiologies lead to WMLs. However, there is no specific therapy or effective drugs for relieving WMLs. Natural products and their derivatives originate from bacterial, fungal, plant, and marine animal sources, many of which have multiple therapeutic targets. Compared to single target compounds, natural products and their derivatives are promising to be developed as better drugs to attenuate WMLs. Thus, this review attempts to summarize the status of natural products and their derivatives (2010-to date) alleviating cerebral white matter lesions for the discovery of new drugs.


Asunto(s)
Productos Biológicos , Sustancia Blanca , Animales , Sustancia Blanca/patología , Productos Biológicos/farmacología
3.
Phytochemistry ; 219: 113964, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38184162

RESUMEN

Six pairs of enantiomeric dilignans, (+)/(-)-magdiligols A-F, have been isolated from an ethanolic extract of the barks of Magnolia officinalis var. biloba. Their chemical structures were elucidated by extensive spectroscopic analyses, NMR calculation with DP4+ analysis, and the electronic circular dichroism spectra calculation. (+)/(-)-1-3 possessed a dihydrobenzopyran ring, while a propyl chain of 1 was linked via ether bond. (+)/(-)-Magdiligols D and E ((+)/(-)-4 and 5) were dilignans possessing a furan ring. (+)-Magdiligol B ((+)/(-)-2), (+)/(-)-magdiligol C ((+)/(-)-3), and racemes 2, 3, and 5 showed potential hepatoprotective effects against APAP-induced HepG2 cell damage, increased the cell viability from 65.4% to 72.7, 78.7.76.6, 73.9, 77.9 and 73.2%, via decreasing the level of the live enzymes ALH and LDH consistently. (+)/(-)-Magdiligols B-D ((+)/(-)-2-4) and (+)/(-)-magdiligol F ((+)/(-)-6) exhibited significant antioxidative activity. (+)/(-)-Magdiligols B-C ((+)/(-)-2 and 3), (-)-magdiligol D ((-)-4), and (+)-magdiligol E ((+)-5) displayed significant PTP1B inhibitory activity with IC50 values 1.41-3.42 µM. (+)/(-)-Magdiligol B ((+)/(-)-2), and its raceme (2) demonstrated α-glucosidase inhibitory activity with the IC50 values 1.47, 2.88 and 1.85 µM, respectively.


Asunto(s)
Magnolia , Humanos , Magnolia/química , Espectroscopía de Resonancia Magnética , Células Hep G2 , Estructura Molecular
4.
Food Funct ; 15(2): 1052, 2024 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-38205631

RESUMEN

Correction for 'Gardenia jasminoides J. Ellis extract alleviated white matter damage through promoting the differentiation of oligodendrocyte precursor cells via suppressing neuroinflammation' by Caixia Zang et al., Food Funct., 2022, 13, 2131-2141, https://doi.org/10.1039/D1FO02127C.

5.
Int J Biol Macromol ; 248: 125895, 2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37481185

RESUMEN

This study identified two homogeneous acidic polysaccharides from Gardeniae fructus, GJP50-3 and GJP50-4, which exhibited potential immunomodulatory activities in macrophage activation assays, via liquid-chip technology, and in a zebrafish model. Monosaccharide composition analysis and gel permeation chromatography revealed that GJP50-3 and GJP50-4 were composed of Rha, GalA, Glc, Gal, and Ara in specific ratios and had molecular weights of 91.5 kDa and 140.3 kDa, respectively. Based on FT-IR, GC-MS, and NMR analyses, these polysaccharides were identified as typical pectin polysaccharides with methylation degrees of 24.7 % and 21.4 %, respectively. The primary structures of GJP50-3 and GJP50-4 included linear HG domains and branched RG-I domains with arabinans and AG side chains. In vitro, GJP50-3 and GJP50-4 could stimulate NO release and increase the secretion of TNF-α in a RAW 264.7 macrophage model. Luminex liquid suspension chip detection revealed that GJP50-3 significantly promoted the secretion of multiple interleukins [IL-6, IL-9, IL-10, IL-12 (p40), IL-12 (p70), IL-13], TNF-α, and chemokines (G-CSF, GM-CSF, MCP-1 and RANTES). In vivo, these polysaccharides could also increase NO release and neutrophil count in a zebrafish model. These findings suggested that GJP50-3 and GJP50-4 might have the potential to be used as immunomodulators in the food and pharmaceutical industries.


Asunto(s)
Gardenia , Animales , Pez Cebra , Factor de Necrosis Tumoral alfa , Espectroscopía Infrarroja por Transformada de Fourier , Polisacáridos/farmacología , Polisacáridos/química , Interleucina-12
6.
Signal Transduct Target Ther ; 8(1): 101, 2023 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-36894540

RESUMEN

Tutin, an established toxic natural product that causes epilepsy in rodents, is often used as a tool to develop animal model of acute epileptic seizures. However, the molecular target and toxic mechanism of tutin were unclear. In this study, for the first time, we conducted experiments to clarify the targets in tutin-induced epilepsy using thermal proteome profiling. Our studies showed that calcineurin (CN) was a target of tutin, and that tutin activated CN, leading to seizures. Binding site studies further established that tutin bound within the active site of CN catalytic subunit. CN inhibitor and calcineurin A (CNA) knockdown experiments in vivo proved that tutin induced epilepsy by activating CN, and produced obvious nerve damage. Together, these findings revealed that tutin caused epileptic seizures by activating CN. Moreover, further mechanism studies found that N-methyl-D-aspartate (NMDA) receptors, gamma-aminobutyric acid (GABA) receptors and voltage- and Ca2+- activated K+ (BK) channels might be involved in related signaling pathways. Our study fully explains the convulsive mechanism of tutin, which provides new ideas for epilepsy treatment and drug development.


Asunto(s)
Calcineurina , Epilepsia , Animales , Ratones , Calcineurina/genética , Calcineurina/metabolismo , Epilepsia/inducido químicamente , Epilepsia/tratamiento farmacológico , Epilepsia/genética , Picrotoxina , Receptores de GABA/metabolismo , Receptores de N-Metil-D-Aspartato , Convulsiones/inducido químicamente , Convulsiones/genética
7.
J Ethnopharmacol ; 310: 116292, 2023 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-36931412

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Yinma Jiedu Granule (YMJD) is a traditional Chinese patent medicine (CPM), which has been proved to have anti-inflammatory effects and therapeutical effects on obstructive pulmonary disease. AIM OF STUDY: The purpose of the current investigation is to find out if YMJD can alleviate acute lung injury (ALI) induced by lipopolysaccharide (LPS) in rats and its underlying mechanisms. MATERIALS AND METHODS: Rats were treated with either vehicle or YMJD for 14 consecutive days, and 2 h after the last administration, the rat model of ALI was induced by the intratracheal instillation of LPS. High performance liquid chromatography (HPLC) was applied for the fingerprint analysis of YMJD. The efficacy and molecular mechanisms were investigated. RESULTS: The results showed that treatment with YMJD improved the general state of rats, reduced weight loss and serum lactate (LA) levels, attenuated pulmonary edema and pathological damage of the lung tissue. Moreover, we found that YMJD effectively decreased the infiltration of white blood cells (WBC), lymphocytes (LYM), mononuclear cells (MON) and neutrophils (NEUT) in bronchoalveolar lavage fluid (BALF), reduced the concentration of tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) and inhibited inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) expression in the lung tissue. Additionally, we found that YMJD could significantly increase the activity of superoxide dismutase (SOD) and reduce the malondialdehyde (MDA) level in the lung tissue. By employing RNA-sequencing, we have identified that JAK2/STAT1 is an important pathway that is involved in the lung protection of YMJD, and further Western blot assay verified that YMJD could effectively inhibit the activation of the JAK2/STAT1 pathway. CONCLUSIONS: YMJD could attenuate LPS-induced ALI through suppressing inflammation and oxidative stress in the lung tissue of rats, associating with the inhibition of JAK2/STAT1 activation. These findings provide evidence for the clinical use of YMJD for treatment of inflammatory pulmonary diseases like ALI.


Asunto(s)
Lesión Pulmonar Aguda , Edema Pulmonar , Ratas , Animales , Lipopolisacáridos/toxicidad , Lipopolisacáridos/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/metabolismo , Pulmón , Inflamación/patología , Edema Pulmonar/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
8.
Neurobiol Dis ; 177: 105988, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36603746

RESUMEN

CXC chemokine receptor 2 (CXCR2) plays an important role in demyelinating diseases, but the detailed mechanisms were not yet clarified. In the present study, we mainly investigated the critical function and the potential molecular mechanisms of CXCR2 on oligodendrocyte precursor cell (OPC) differentiation and remyelination. The present study demonstrated that inhibiting CXCR2 significantly enhanced OPC differentiation and remyelination in primary cultured OPCs and ethidium bromide (EB)-intoxicated rats by facilitating the formation of myelin proteins, including PDGFRα, MBP, MAG, MOG, and Caspr. Further investigation identified phosphodiesterase 10A (PDE10A) as a main downstream protein of CXCR2, interacting with the receptor to regulate OPC differentiation, in that inhibition of CXCR2 reduced PDE10A expression while suppression of PDE10A did not affect CXCR2. Furthermore, inhibition of PDE10A promoted OPC differentiation, whereas overexpression of PDE10A down-regulated OPC differentiation. Our data also revealed that inhibition of CXCR2/PDE10A activated the cAMP/ERK1/2 signaling pathway, and up-regulated the expression of key transcription factors, including SOX10, OLIG2, MYRF, and ZFP24, that ultimately promoted remyelination and myelin protein biosynthesis. In conclusion, our findings suggested that inhibition of CXCR2 promoted OPC differentiation and enhanced remyelination by regulating PDE10A/cAMP/ERK1/2 signaling pathway. The present data also highlighted that CXCR2 may serve as a potential target for the treatment of demyelination diseases.


Asunto(s)
Enfermedades Desmielinizantes , Remielinización , Ratas , Animales , Ratones , Remielinización/fisiología , Receptores de Interleucina-8B/metabolismo , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/tratamiento farmacológico , Enfermedades Desmielinizantes/metabolismo , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Transducción de Señal , Diferenciación Celular/fisiología , Ratones Endogámicos C57BL , Hidrolasas Diéster Fosfóricas/metabolismo
9.
Am J Chin Med ; 51(1): 53-72, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36458485

RESUMEN

Endoplasmic reticulum stress (ERS) is involved in the pathological process of vascular dementia (VD). GJ-4 is extracted from Gardenia jasminoides J. Ellis and has been reported to have protective roles in ischemia-related brain damage. However, the role of GJ-4 in ERS has not been elucidated. We established a VD rat model through bilateral common carotid arteries occlusion (2-VO). The rats were intragastrically administrated with GJ-4 (10, 25, and 50[Formula: see text]mg/kg) and nimodipine (10[Formula: see text]mg/kg). Data from a Morris water maze test showed that GJ-4 could significantly alleviate learning and memory deficits in VD rats. Nissl and cleaved caspase-3 staining revealed that GJ-4 can inhibit apoptosis and thus exert a protective role in the brain of 2-VO rats. Western blot results suggested that GJ-4 significantly reduced ERS-related protein expression and inhibited apoptosis through suppression of the PERK/eIF2[Formula: see text]/ATF4/CHOP signaling pathway. For in vitro studies, the oxygen-glucose deprivation (OGD) SH-SY5Y model was employed. Western blot and Hoechst 33342/PI double staining were utilized to explore the effects of crocetin, the main active metabolite of GJ-4. Like GJ-4 in vivo, crocetin in vitro also decreased ERS-related protein expression and inhibited the activation of the PERK/eIF2[Formula: see text]/ATF4/CHOP signaling pathway. Thus, crocetin exerted similar protective roles on OGD challenged SH-SY5Y cells in vitro. In summary, GJ-4 and crocetin reduce the ERS in the brain of VD rats and SY5Y cells subjected to OGD and inhibit neuronal apoptosis through suppression of the PERK/eIF2[Formula: see text]/ATF4/CHOP pathway, suggesting that GJ-4 may be useful for the treatment of VD.


Asunto(s)
Demencia Vascular , Gardenia , Neuroblastoma , Ratas , Humanos , Animales , Demencia Vascular/tratamiento farmacológico , Demencia Vascular/etiología , Factor 2 Eucariótico de Iniciación/farmacología , Apoptosis , Estrés del Retículo Endoplásmico
10.
Bioorg Chem ; 129: 106179, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36244322

RESUMEN

A series of novel pyranocarbazole alkaloids were designed and synthesized as derivatives of Claulansine F and CZ-7. Some of the compounds showed strong neuroprotective effects and anti-lipid peroxidation capacity. Among these compounds, 10b, introduced leucine at the C-3 position of pyranocarbazole, was the most active in inhibiting the programmed death of SH-SY5Y cells. This compound exhibited stronger free radical scavenging activity than Edaravone. Furthermore, 10b could penetrate the blood-brain barrier (BBB). More importantly, 10b showed a tendency of improvement in learning and memory in the dose range of 10-40 mg/kg. The research on mechanisms indicated that 10b could reduce oxidative stress in the brain of Aß25-35-intoxicated mice, and then improve the cognitive function of Aß25-35-intoxicated mice. Our findings suggest that 10b may be promising for further evaluation as an intervention for Alzheimer's Disease.


Asunto(s)
Enfermedad de Alzheimer , Antioxidantes , Cognición , Diseño de Fármacos , Fármacos Neuroprotectores , Animales , Humanos , Ratones , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides , Antioxidantes/síntesis química , Antioxidantes/química , Antioxidantes/farmacología , Cognición/efectos de los fármacos , Fármacos Neuroprotectores/síntesis química , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/farmacología , Línea Celular Tumoral , Estrés Oxidativo/efectos de los fármacos
11.
Food Nutr Res ; 662022.
Artículo en Inglés | MEDLINE | ID: mdl-35950104

RESUMEN

Background: GJ-4 is extracted from Gardenia jasminoides J. Ellis (Fructus Gardenia) with crocin composition and has been demonstrated to improve memory deficits in several dementia models in our previous studies. Objective: This study aimed to evaluate the effects of GJ-4 on hyperlipidemic vascular dementia (VD) and explore the underlying mechanisms. Design: In the current study, we employed a chronic hyperlipidemic VD rat model by permanent bilateral common carotid arteries occlusion (2-VO) based on high-fat diet (HFD), which is an ideal model to mimic the clinical pathogenesis of human VD. Results: Our results showed that GJ-4 could significantly reduce serum lipids level and improve cerebral blood flow in hyperlipidemic VD rats. Additionally, treatment with GJ-4 remarkedly ameliorated memory impairment and alleviated neuronal injury. Mechanistic investigation revealed that the neuroprotective effects of GJ-4 might be attributed to the inhibition of microglia-mediated neuro-inflammation via regulating the M1/M2 polarization. Our data further illustrated that GJ-4 could regulate the phenotype of microglia through activating the peroxisome proliferator-activated receptor-γ (PPAR-γ) and subsequently inhibited nuclear factor-κB (NF-κB) nuclear translocation and increased CCAAT/enhancer-binding protein ß (C/EBPß) expression. Conclusion: Our results implied that GJ-4 might be a promising drug to improve VD through the regulation of microglial M1/M2 polarization and the subsequent inhibition of neuro-inflammation.

12.
Inflammation ; 45(6): 2375-2387, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35917097

RESUMEN

Dual-specificity tyrosine phosphorylation-regulated kinase 1A (Dyrk1A) is a highly conserved protein kinase, playing a key role in the regulation of physiological brain functions and pathological processes. In Alzheimer's disease (AD), Dyrk1A promotes hyperphosphorylation of tau protein and abnormal aggregation of amyloid-ß protein (Aß). This study investigated the role of Dyrk1A in regulating neuroinflammation, another critical factor that contributes to AD. In the present study, we used an immortalized murine BV2 microglia cell line induced by lipopolysaccharide (LPS) to study neuroinflammation. The expression and activity of Dyrk1A kinase were both increased by inflammation. Dyrk1A inhibition using harmine or siRNA silencing significantly reduced the production of proinflammatory factors in LPS-stimulated BV2 cells. Reactive oxygen species (ROS), tumor necrosis factor-α (TNF-α), and nitric oxide (NO), as well as the expression of the inflammatory proteins, cyclooxygenase 2 (COX2), and inducible nitric synthase (iNOS), were attenuated. In vivo, in ICR mice injected with LPS into the left lateral cerebral ventricle, harmine (20 mg/kg) administration decreased the expression of inflammatory proteins in the cortex and hippocampus of mice brain. In addition, immunohistochemical detection of ionized calcium-binding adapter molecule 1 (Iba1) and Nissl staining showed that harmine significantly attenuated microglia activation and neuronal damage in the CA1 region of hippocampus. Further mechanistic studies indicated that Dyrk1A suppression may be related to inhibition of the TLR4/NF-κB signaling pathway in LPS-induced neuroinflammation. Taken together, our studies suggest that Dyrk1A may be a novel target for the treatment of neurodegenerative diseases with an inflammatory component.


Asunto(s)
Enfermedades Neuroinflamatorias , Proteínas Serina-Treonina Quinasas , Animales , Ratones , Harmina , Lipopolisacáridos , Ratones Endogámicos ICR , Microglía/metabolismo , Enfermedades Neuroinflamatorias/tratamiento farmacológico , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Transducción de Señal , Receptor Toll-Like 4/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Quinasas DyrK
13.
Food Funct ; 13(4): 2131-2141, 2022 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-35112688

RESUMEN

Increasing evidence has highlighted the role of white matter damage in the pathology of Alzheimer's disease (AD). Previous research has shown that a mixture of crocin analogues (GJ-4), Gardenia jasminoides J. Ellis extract, improved cognition in several AD mouse models, but the mechanism remains unclear. The aim of the present study was to investigate the effects and underlying mechanisms of GJ-4 on white matter damage. Proteomic analysis and western blotting results suggested that the level of myelin-related proteins, including myelin basic protein (MBP), myelin associated glycoprotein (MAG) and myelin associated oligodendrocyte basic protein (MOBP), was significantly decreased in the brain of PrP-hAßPPswe/PS1ΔE9 (APP/PS1) transgenic mice, and GJ-4 treatment increased the expressions of these proteins. This result revealed that GJ-4 could ameliorate myelin injury, suggesting that this might be a possible mechanism of GJ-4 on cognition. To validate the effects of GJ-4 on myelin, a metabolite of GJ-4, crocetin, which can pass through the blood-brain barrier, was applied in in vitro experiments. A mechanistic study revealed that crocetin significantly promoted the differentiation of primary cultured oligodendrocyte precursor cells to oligodendrocytes through up-regulation of nuclear Ki67 and transcription factor 2 (Olig2). Oligodendrocytes, the myelin-forming cells, have been reported to be lifelong partners of neurons. Therefore, to investigate the effects of crocetin on myelin and neurons, lysophosphatidylcholine (LPC)-treated primary mixed midbrain neuronal/glial culture was used. Immunofluorescence results indicated that crocetin treatment protected neurons and suppressed microglial activation against LPC-induced injury. To further discern the effects of GJ-4 on white matter injury and neuroinflammation, an LPC-induced mouse model was developed. GJ-4 administration increased oligodendrocyte proliferation, differentiation, and myelin repair. The mechanistic study indicated that GJ-4 improved white matter injury through the regulation of neuroinflammatory dysfunction. These data indicated that GJ-4 effectively repaired white matter damage in the LPC-treated mice. Thus, the present study supported GJ-4 as a potential therapeutic agent for AD and white matter related diseases.


Asunto(s)
Gardenia , Fármacos Neuroprotectores/farmacología , Extractos Vegetales/farmacología , Enfermedad de Alzheimer/prevención & control , Animales , Modelos Animales de Enfermedad , Humanos , Lisofosfatidilcolinas , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Transgénicos , Proteína Básica de Mielina/metabolismo , Enfermedades Neuroinflamatorias/inducido químicamente , Enfermedades Neuroinflamatorias/prevención & control , Fármacos Neuroprotectores/uso terapéutico , Oligodendroglía/efectos de los fármacos , Fitoterapia , Extractos Vegetales/uso terapéutico , Proteómica , Sustancia Blanca/efectos de los fármacos
14.
Acta Pharmacol Sin ; 43(2): 285-294, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34593974

RESUMEN

Neuroinflammation plays an important role in neurodegenerative diseases, such as Parkinson's disease (PD) and Alzheimer's disease. HACE1 (HECT domain and Ankyrin repeat Containing E3 ubiquitin-protein ligase 1) is a tumor suppressor. Recent evidence suggests that HACE1 may be involved in oxidative stress responses. Due to the critical role of ROS in neuroinflammation, we speculated that HACE1 might participate in neuroinflammation and related neurodegenerative diseases, such as PD. In this study, we investigated the role of HACE1 in neuroinflammation of PD models. We showed that HACE1 knockdown exacerbated LPS-induced neuroinflammation in BV2 microglial cells in vitro through suppressing ubiquitination and degradation of activated Rac1, an NADPH oxidase subunit. Furthermore, we showed that HACE1 exerted vital neuronal protection through increasing Rac1 activity and stability in LPS-treated SH-SY5Y cells, as HACE1 knockdown leading to lower tolerance to LPS challenge. In MPTP-induced acute PD mouse model, HACE1 knockdown exacerbated motor deficits by activating Rac1. Finally, mutant α-synuclein (A53T)-overexpressing mice, a chronic PD mouse model, exhibited age-dependent reduction of HACE1 levels in the midbrain and striatum, implicating that HACE1 participated in PD pathological progression. This study for the first time demonstrates that HACE1 is a negative regulator of neuroinflammation and involved in the PD pathogenesis by regulating Rac1 activity. The data support HACE1 as a potential target for PD and other neurodegenerative diseases.


Asunto(s)
Trastornos Parkinsonianos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Western Blotting , Línea Celular Tumoral , Técnica del Anticuerpo Fluorescente , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedades Neuroinflamatorias/metabolismo , Prueba de Desempeño de Rotación con Aceleración Constante , Ubiquitinación
15.
Biochem Pharmacol ; 197: 114891, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34968482

RESUMEN

Mitochondria are the principal sites of energy metabolism and provide most of the energy needed for normal cellular function. They are dynamic organelles that constantly undergo fission, fusion and mitophagy to maintain their homeostasis and function. However, dysregulated mitochondrial dynamics and mitophagy leads to reduced ATP generation and mutation of their DNA, which ultimately leads to cell death. Increasing evidence has shown that the FUN14 domain-containing protein 1 (FUNDC1), a novel mitophagy receptor, participates in the process of mitochondrial dynamics and mitophagy and plays a critical role in various human diseases. Herein, we review the role of FUNDC1 in mitophagy and mitochondrial dynamics, thus providing a better understanding of the relationship between the two processes. Moreover, we summarize the treatments targeting FUNDC1, and suggest that FUNDC1 may represent a promising therapeutic target for the treatment of several human diseases such as cardiovascular diseases, metabolic syndrome, cancer and chronic obstructive pulmonary disease (COPD).


Asunto(s)
Proteínas de la Membrana/fisiología , Dinámicas Mitocondriales/fisiología , Proteínas Mitocondriales/fisiología , Mitofagia/fisiología , Animales , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Humanos , Proteínas de la Membrana/química , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/patología , Mitocondrias/metabolismo , Mitocondrias/patología , Proteínas Mitocondriales/química , Neoplasias/metabolismo , Neoplasias/patología
16.
Phytomedicine ; 93: 153780, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34607163

RESUMEN

BACKGROUND: Accumulating evidence demonstrates that traditional Chinese medicines that act on multiple targets could effectively treat various multi-etiological diseases, including cerebrovascular diseases, Alzheimer's disease (AD), Parkinson's disease (PD) and so on. Previous studies have shown that crocin richments (GJ-4), Gardenia jasminoides J.Ellis extract, provide neuroprotective effects on cognitive impairments in AD mouse models. However, the mechanism how GJ-4 improves cognition remains still unclear. PURPOSE: The aim of this study was to uncover the protective effects and underlying mechanism of GJ-4 on PrP-hAßPPswe/PS1ΔE9 (APP/PS1) transgenic mice. METHODS: APP/PS1 mice were given GJ-4 (10, 20, and 50 mg/kg), donepezil (5 mg/kg) and memantine (5 mg/kg) orally at eight months of age for 12 consecutive weeks. Morris water maze and novel object recognition were conducted to assess the cognitive ability of mice. The release of inflammatory cytokines was determined by RT-PCR assay, and the pathological features of neurons and microglia were assayed by immunohistochemistry and immunofluorescence assay. The expression of Aß-related proteins and signaling pathways were determined by Western blot. RESULTS: The behavioral results revealed that GJ-4 ameliorated the cognitive deficits of APP/PS1 mice measured by Morris water maze and novel object recognition tests. Mechanism studies indicated that GJ-4 significantly decreased ß-amyloid (Aß) level through reducing Aß production and promoting Aß degradation. It has been reported that Aß plaques trigger the hyper-phosphorylation of tau protein in APP/PS1 mice. Consistent with previous studies, hyper-phosphorylation of tau was also occurred in APP/PS1 mice in the present study, and GJ-4 inhibited Tau phosphorylation at different sites. Overwhelming evidence indicates that neuroinflammation stimulated by Aß and hyperphosphorylated tau is involved in the pathological progression of AD. We found that GJ-4 suppressed neuroinflammatory responses in the brain through regulating phosphatidylinositide 3-kinase/AKT (PI3K/AKT) signaling pathway activation, and subsequent expression of inflammatory proteins and release of inflammatory cytokines. CONCLUSION: Altogether, GJ-4 ameliorated cognition of APP/PS1 transgenic mice through multiple targets, including Aß, tau and neuroinflammation. This study provides a solid research basis for further development of GJ-4 as a potential candidate for the treatment of AD.


Asunto(s)
Disfunción Cognitiva , Medicamentos Herbarios Chinos , Gardenia , Animales , Cognición , Disfunción Cognitiva/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Ratones , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas
17.
Mol Neurobiol ; 58(11): 5743-5755, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34398403

RESUMEN

Microglial activation-induced neuroinflammation is critical in the pathogenesis of neurodegenerative diseases. Activated microglia are regulated mainly by innate pattern recognition receptors (PRRs) on their surface, of which macrophage receptor with collagenous structure (Marco) is a well-characterized scavenger receptor constitutively expressed on specific subsets of macrophages, including microglia. Increasing evidence has shown that Marco is involved in the pathogenesis of a range of inflammatory processes. However, research on the role of Marco in regulating neuroinflammation has reported conflicting results. In the present study, we examined the role Marco played in triggering neuroinflammation and its underlying mechanisms. The results demonstrated that silencing the Marco gene resulted in a significantly reduced neuroinflammatory response and vice versa. α-Syn stimulation in Marco overexpressing cells induced a pronounced inflammatory response, suggesting that Marco alone could trigger an inflammatory response. We also found that TLR2 significantly promoted Marco-mediated neuroinflammation, indicating TLR2 was an important co-receptor of Marco. Knocking down the TLR2 gene in microglia and mouse substantia nigra resulted in decreased expression of Marco. Subsequent mechanistic studies showed that deleting the SRCR domain of Marco resulted in disruption of the inflammatory response and the interaction between TLR2 and Marco. This suggested that TLR2 binds directly to the SRCR domain of Marco and regulates Marco-mediated neuroinflammation. In summary, this investigation revealed that TLR2 could potentiate Marco-mediated neuroinflammation by interacting with the SRCR domain of Marco, providing a new target for inhibiting neuroinflammation in neurodegenerative diseases.


Asunto(s)
Enfermedades Neuroinflamatorias/metabolismo , Receptores Inmunológicos/metabolismo , Receptor Toll-Like 2/metabolismo , Animales , Línea Celular , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía , Óxido Nítrico/metabolismo , Polisacáridos/farmacología , Unión Proteica , Dominios Proteicos , Mapeo de Interacción de Proteínas , Interferencia de ARN , Receptores Inmunológicos/antagonistas & inhibidores , Receptores Inmunológicos/química , Proteínas Recombinantes/metabolismo , Receptor Toll-Like 2/antagonistas & inhibidores , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética , alfa-Sinucleína/farmacología
18.
Acta Pharm Sin B ; 11(5): 1213-1226, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-34094829

RESUMEN

The gut microbiota plays an important role in regulating the pharmacokinetics and pharmacodynamics of many drugs. FLZ, a novel squamosamide derivative, has been shown to have neuroprotective effects on experimental Parkinson's disease (PD) models. FLZ is under phase Ⅰ clinical trial now, while the underlying mechanisms contributing to the absorption of FLZ are still not fully elucidated. Due to the main metabolite of FLZ was abundant in feces but rare in urine and bile of mice, we focused on the gut microbiota to address how FLZ was metabolized and absorbed. In vitro studies revealed that FLZ could be exclusively metabolized to its major metabolite M1 by the lanosterol 14 alpha-demethylase (CYP51) in the gut microbiota, but was almost not metabolized by any other metabolism-related organs, such as liver, kidney, and small intestine. M1 was quickly absorbed into the blood and then remethylated to FLZ by catechol O-methyltransferase (COMT). Notably, dysbacteriosis reduced the therapeutic efficacy of FLZ on the PD mouse model by inhibiting its absorption. The results show that the gut microbiota mediate the absorption of FLZ through a FLZ-M1-FLZ circulation. Our research elucidates the vital role of the gut microbiota in the absorption of FLZ and provides a theoretical basis for clinical pharmacokinetic studies and clinical application of FLZ in the treatment of PD.

19.
Front Pharmacol ; 12: 588003, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33815098

RESUMEN

Mitochondrial dysfunction is involved in the pathogenesis of Parkinson's disease (PD). Mitochondrial morphology is dynamic and precisely regulated by mitochondrial fission and fusion machinery. Aberrant mitochondrial fragmentation, which can result in cell death, is controlled by the mitochondrial fission protein, dynamin-related protein 1 (Drp1). Our previous results demonstrated that FLZ could correct mitochondrial dysfunction, but the effect of FLZ on mitochondrial dynamics remain uncharacterized. In this study, we investigated the effect of FLZ and the role of Drp1 on 1-methyl-4-phenylpyridinium (MPP+)-induced mitochondrial fission in neurons. We observed that FLZ blocked Drp1, inhibited Drp1 enzyme activity, and reduced excessive mitochondrial fission in cultured neurons. Furthermore, by inhibiting mitochondrial fission and ROS production, FLZ improved mitochondrial integrity and membrane potential, resulting in neuroprotection. FLZ curtailed the reduction of synaptic branches of primary cultured dopaminergic neurons caused by MPP+ exposure, reduced abnormal fission, restored normal mitochondrial distribution in neurons, and exhibited protective effects on dopaminergic neurons. The in vitro research results were validated using an MPTP-induced PD mouse model. The in vivo results revealed that FLZ significantly reduced the mitochondrial translocation of Drp1 in the midbrain of PD mice, which, in turn, reduced the mitochondrial fragmentation in mouse substantia nigra neurons. FLZ also protected dopaminergic neurons in PD mice and increased the dopamine content in the striatum, which improved the motor coordination ability of the mice. These findings elucidate this newly discovered mechanism through which FLZ produces neuroprotection in PD.

20.
J Ethnopharmacol ; 267: 113491, 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33091490

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Gardenia jasminoides J. Ellis (Fructus Gardenia) is a traditional Chinese medicine with diverse pharmacological functions, such as anti-inflammation, anti-depression, as well as improvement of cognition and ischemia brain injury. GJ-4 is a natural extract from Gardenia jasminoides J. Ellis (Fructus Gardenia) and has been proved to improve memory impairment in Alzheimer's disease (AD) mouse model in our previous studies. AIM OF THE STUDY: This study aimed to evaluate the therapeutic effects of GJ-4 on vascular dementia (VD) and explore the potential mechanisms. MATERIAL AND METHODS: In our experiment, a focal cerebral ischemia and reperfusion rat model was successfully developed by the middle cerebral artery occlusion and reperfusion (MCAO/R). GJ-4 (10 mg/kg, 25 mg/kg, 50 mg/kg) and nimodipine (10 mg/kg) were orally administered to rats once a day for consecutive 12 days. Learning and memory behavioral performance was assayed by step-down test and Morris water maze test. The neurological scoring test was performed to evaluate the neurological function of rats. 2,3,5-Triphenyltetrazolium chloride (TTC) staining and Nissl staining were respectively employed to determine the infarct condition and neuronal injury of the brain. Iba1 immunohistochemistry was used to show the activation of microglia. Moreover, the synaptic damage and inflammatory level were detected by Western blot. RESULTS: GJ-4 could significantly improve memory impairment, cerebral infraction, as well as neurological deficits of VD rats induced by MCAO/R. Further research indicated VD-induced neuronal injury was alleviated by GJ-4. In addition, GJ-4 could protect synapse of VD rats by upregulating synaptophysin (SYP) expression, post synaptic density 95 protein (PSD95) expression, and downregulating N-Methyl-D-Aspartate receptor 1 (NMDAR1) expression. Subsequent investigation of the underlying mechanisms identified that GJ-4 could suppress neuroinflammatory responses, supported by inhibited activation of microglia and reduced expression of inflammatory proteins, which ultimately exerted neuroprotective effects on VD. Further mechanistic study indicated that janus kinase 2 (JAK2)/signal transducer and activator of transcription 1 (STAT1) pathway was inhibited by GJ-4 treatment. CONCLUSION: These results suggested that GJ-4 might serve as a potential drug to improve VD. In addition, our study indicated that inhibition of neuroinflammation might be a promising target to treat VD.


Asunto(s)
Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Demencia Vascular/prevención & control , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Janus Quinasa 2/metabolismo , Trastornos de la Memoria/prevención & control , Memoria/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Nootrópicos/farmacología , Extractos Vegetales/farmacología , Daño por Reperfusión/prevención & control , Factor de Transcripción STAT1/metabolismo , Animales , Encéfalo/enzimología , Encéfalo/patología , Encéfalo/fisiopatología , Demencia Vascular/enzimología , Demencia Vascular/etiología , Demencia Vascular/psicología , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/farmacología , Gardenia , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/enzimología , Infarto de la Arteria Cerebral Media/fisiopatología , Mediadores de Inflamación/metabolismo , Masculino , Trastornos de la Memoria/enzimología , Trastornos de la Memoria/etiología , Trastornos de la Memoria/psicología , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , Ratas Sprague-Dawley , Daño por Reperfusión/enzimología , Daño por Reperfusión/etiología , Daño por Reperfusión/fisiopatología , Transducción de Señal , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Sinapsis/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...