Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Inflammopharmacology ; 32(1): 393-404, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37429999

RESUMEN

Medulla Tetrapanacis (MT) is a commonly used herb to promote lactation and manage mastitis in lactating mothers. However, its anti-inflammatory and anti-bacterial effects are currently unknown. We hypothesized that MT water extract possesses anti-inflammatory and anti-bacterial effects by modulating macrophage polarization to reduce the release of inflammatory mediators and phagocytosis via inactivation of MAPKs pathways. The chemical composition of the MT water extract was analyzed by UPLC-Orbitrap-mass spectrometry. The anti-inflammatory and anti-bacterial properties of the MT water extract were examined using LPS-stimulated inflammation and Staphylococcus aureus infection model in RAW 264.7 cells, respectively. The underlying mechanism of action of the MT water extract was also investigated. We identified eight compounds by UPLC-Orbitrap-mass spectrometry that are abundant within the MT water extract. MT water extract significantly suppressed LPS-induced nitric oxide, TNF-α and IL-6 secretion in RAW 264.7 cells which was accompanied by the promotion of macrophage polarization from pro-inflammatory towards anti-inflammatory phenotypes. MT water extract significantly suppressed the LPS-induced MAPK activation. Finally, MT water extract decreased the phagocytic capacity of the RAW 264.7 cells against S. aureus infection. MT water extract could suppress LPS-induced inflammation by promoting macrophages towards an anti-inflammatory phenotype. In addition, MT also inhibited the growth of S. aureus.


Asunto(s)
Lactancia , Lipopolisacáridos , Femenino , Humanos , Lipopolisacáridos/farmacología , Staphylococcus aureus , Transducción de Señal , Inflamación/tratamiento farmacológico , Macrófagos , Antiinflamatorios/farmacología
2.
Phytother Res ; 37(10): 4442-4456, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37259475

RESUMEN

Cancer metastasis remains the most common cause of death in breast cancer patients. Tumor-associated macrophages (TAMs) are a novel therapeutic target for the treatment of metastatic breast cancer. Despite the good anti-cancer activity of garcinone E (GE), there are no reports on its therapeutic effects on breast cancer metastasis. The objective of this study was to examine the anti-cancer effects of GE on metastatic breast cancer. RAW 264.7 and THP-1 cells were polarized to M2 macrophages by IL-4/IL-13 in vitro. A 4T1 mouse breast cancer model and the tail vein breast cancer metastasis model were used to explore the effect of GE on breast cancer growth and metastasis in vivo. In vitro studies showed that GE dose-dependently suppressed IL-4 + IL-13-induced expression of CD206 in both RAW 264.7 cells and differentiated THP-1 macrophages. However, GE did not affect the LPS + IFN-γ-induced polarization to the M1-like macrophages in vitro. GE inhibited the expression of the M2 macrophage specific genes in RAW 264.7 cells, and simultaneously impaired M2 macrophage-induced breast cancer cell proliferation and migration, and angiogenesis. In animal studies, GE significantly suppressed tumor growth, angiogenesis, and lung metastasis in 4T1 tumor-bearing mice, without causing toxicity. In both tumor and lung tissues, the proportion of M2-like TAMs was significantly decreased while the proportion of M1-like TAMs was markedly increased by GE treatment. Mechanistically, GE inhibited phosphorylation of STAT6 in vitro and in vivo. Our results demonstrate for the first time that GE suppresses breast cancer growth and pulmonary metastasis by modulating M2-like macrophage polarization through the STAT6 signaling pathway.


Asunto(s)
Neoplasias de la Mama , Humanos , Animales , Ratones , Femenino , Neoplasias de la Mama/patología , Macrófagos Asociados a Tumores , Línea Celular Tumoral , Interleucina-4/metabolismo , Interleucina-4/farmacología , Interleucina-4/uso terapéutico , Interleucina-13/metabolismo , Interleucina-13/farmacología , Interleucina-13/uso terapéutico , Transducción de Señal , Factor de Transcripción STAT6/metabolismo , Factor de Transcripción STAT6/farmacología
3.
Cell Mol Neurobiol ; 42(4): 973-984, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-33392917

RESUMEN

Cerebral ischemia is a cerebrovascular disease with high morbidity and mortality that poses a significant burden on society and the economy. About 60% of cerebral ischemia is caused by thrombus, and the formation of thrombus proceeds from insoluble fibrin, following its transformation from liquid fibrinogen. In thrombus-induced ischemia, increased permeability of the blood-brain barrier (BBB), followed by the extravasation of blood components into the brain results in an altered brain microenvironment. Changes in the brain microenvironment affect brain function and the neurovascular unit (NVU), the working unit of the brain. Recent studies have reported that coagulation factors interact with the NVU and its components, but the specific function of this interaction is highly speculative and warrants further investigations. In this article, we reviewed the role of coagulation factors in cerebral ischemia and the role of coagulation factors in thrombosis. Additionally, the influence of thrombin on the NVU is introduced, as well as in the function of NVU, which may help to explore part of brain injury mechanism during ischemia. Lastly, we propose some novel therapeutic approaches on ischemic stroke by reducing the risk of coagulation.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Barrera Hematoencefálica , Encéfalo/irrigación sanguínea , Humanos , Trombina
4.
Mol Biol Rep ; 49(11): 11201-11208, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36107375

RESUMEN

BACKGROUND: Nucleoside transporters are crucial in regulating the functions of adenosine. This study investigated the contribution of equilibrative nucleoside transporter (ENT) type 4 to adenosine transport in cardiomyocytes under simulated ischemic conditions and whether the inhibition of ENT4 could protect cardiomyocytes against ischemia-reperfusion injury. METHODS: AC16 human cardiomyocytes were used to create a model to simulate ischemia/reperfusion injury. ENT4 activity was inhibited by decynium-22 or specific siRNA against ENT4. The protein expressions of nucleoside transporters were measured by western blot analysis. The transport activity was studied by [3?H]adenosine uptake. The cell injury was studied by biochemical assays. RESULTS: The [3?H]adenosine uptake in AC16 cells was predominantly mediated by ENTs. ENT1 to ENT4 were present in AC16 cells and their protein expression levels were comparable in normal and ischemic conditions. Decynium-22 or siRNA against ENT4 did not affect the adenosine uptake in AC16 cells under normal conditions but could inhibit the adenosine uptake in AC16 cells by 28% under ischemic conditions. In addition, the cell viability and lactate dehydrogenase release of AC16 cells under ischemia conditions could be reduced by decynium-22 or siRNA against ENT4. CONCLUSION: The cell culture model has suggested that ENT4 may play a role in adenosine transport in cardiomyocytes under ischemic conditions. Inhibition or downregulation of ENT4 may be a potential approach for cardioprotection but this notion should be further validated using animal model.


Asunto(s)
Miocitos Cardíacos , Daño por Reperfusión , Animales , Humanos , Miocitos Cardíacos/metabolismo , Adenosina/metabolismo , Nucleósidos/metabolismo , ARN Interferente Pequeño/metabolismo , Daño por Reperfusión/metabolismo , Isquemia
5.
J Neurochem ; 156(2): 249-261, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32891068

RESUMEN

Neuroinflammation is believed to play a primary role in the pathogenesis of most neurodegenerative diseases including Alzheimer's disease, Parkinson's disease and schizophrenia. Currently, suitable in vitro neuroinflammation models for studying cellular interactions and inflammatory mechanisms at the neurovascular unit are still scarce. In this study, we established an experimentally flexible tri-culture neuroinflammation model combining murine microglial cells (N11), mouse neuroblastoma Nuro2A cell lines and brain microvascular endothelial MVEC(B3) cells in a transwell co-culture system stimulated with lipopolysaccharides. Neuroinflammation was induced in this tri-culture model as manifested by activated N11 cells via toll-like receptor 4, resulting in increased release of proinflammatory mediators (nitric oxide, interleukin-6 and tumour necrosis factor-α) through the activation of nuclear factor-κB signalling pathway. The released inflammatory cytokines from N11 in turn, damaged the tight junction in microvascular endothelial MVEC(B3) cells, increased permeability of endothelial barrier, and induced tau phosphorylation and up-regulated caspase-3 expression in mouse neuroblastoma Nuro2A cell lines, leading to neuroinflammation injury. In summary, this tri-culture inflammation model mimics the microenvironment, the cellular crosstalk and the molecular events that take place during neuroinflammation. It provides a robust in vitro model for studying neuroinflammation mechanisms and screening for potential therapeutics to treat various neurodegenerative diseases.


Asunto(s)
Técnicas de Cultivo de Célula , Células Endoteliales , Inflamación , Microglía , Neuronas , Animales , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Ratones
6.
Clin Sci (Lond) ; 134(18): 2521-2534, 2020 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-32936248

RESUMEN

Vitamin D deficiency has been associated with human abdominal aortic aneurysm (AAA); however, its role in AAA pathogenesis is unclear. The aim of the present study was to investigate the effect of vitamin D deficiency on AAA development and examine if administering cholecalciferol (CCF) could limit growth of established AAA within the angiotensin-II (AngII) infused apolipoprotein E-deficient mouse model. Mice were rendered vitamin D deficiency through dietary restriction and during AngII infusion developed larger AAAs as assessed by ultrasound and ex vivo morphometry that ruptured more commonly (48% vs. 19%; P=0.028) than controls. Vitamin D deficiency was associated with increased aortic expression of osteopontin and matrix metalloproteinase-2 and -9 than controls. CCF administration to mice with established aortic aneurysms limited AAA growth as assessed by ultrasound (P<0.001) and ex vivo morphometry (P=0.036) and reduced rupture rate (8% vs. 46%; P=0.031). This effect was associated with up-regulation of circulating and aortic sclerostin. Incubation of human aortic smooth muscle cells with 1,25-dihyroxyvitamin D3 (the active metabolite of vitamin D) for 48 h induced up-regulation of sclerostin (P<0.001) and changed the expression of a range of other genes important in extracellular matrix remodeling. The present study suggests that vitamin D deficiency promotes development of large rupture-prone aortic aneurysms in an experimental model. CCF administration limited both growth and rupture of established aneurysms. These effects of vitamin D appeared to be mediated via changes in genes involved in extracellular matrix remodeling, particularly sclerostin.


Asunto(s)
Aneurisma de la Aorta Abdominal/tratamiento farmacológico , Aneurisma de la Aorta Abdominal/etiología , Rotura de la Aorta/tratamiento farmacológico , Rotura de la Aorta/etiología , Colecalciferol/uso terapéutico , Suplementos Dietéticos , Progresión de la Enfermedad , Deficiencia de Vitamina D/complicaciones , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Angiotensina II , Animales , Aorta Abdominal/efectos de los fármacos , Aorta Abdominal/metabolismo , Aorta Abdominal/patología , Aorta Abdominal/fisiopatología , Aneurisma de la Aorta Abdominal/fisiopatología , Rotura de la Aorta/fisiopatología , Apolipoproteínas E/deficiencia , Presión Sanguínea/efectos de los fármacos , Restricción Calórica , Colecalciferol/farmacología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Deficiencia de Vitamina D/fisiopatología
7.
Clin Sci (Lond) ; 134(9): 1049-1061, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32309850

RESUMEN

Abdominal aortic aneurysm (AAA) is an important cause of mortality in older adults. Chronic inflammation and excessive matrix remodelling are considered important in AAA pathogenesis. Kinins are bioactive peptides important in regulating inflammation. Stimulation of the kinin B2 receptor has been previously reported to promote AAA development and rupture in a mouse model. The endogenous B2 receptor agonist, bradykinin, is generated from the kallikrein-kinin system following activation of plasma kallikrein by Factor XII (FXII). In the current study whole-body FXII deletion, or neutralisation of activated FXII (FXIIa), inhibited expansion of the suprarenal aorta (SRA) of apolipoprotein E-deficient mice in response to angiotensin II (AngII) infusion. FXII deficiency or FXIIa neutralisation led to decreased aortic tumor necrosis factor-α-converting enzyme (TACE/a disintegrin and metalloproteinase-17 (aka tumor necrosis factor-α-converting enzyme) (ADAM-17)) activity, plasma kallikrein concentration, and epithelial growth factor receptor (EGFR) phosphorylation compared with controls. FXII deficiency or neutralisation also reduced Akt1 and Erk1/2 phosphorylation and decreased expression and levels of active matrix metalloproteinase (Mmp)-2 and Mmp-9. The findings suggest that FXII, kallikrein, ADAM-17, and EGFR are important molecular mediators by which AngII induces aneurysm in apolipoprotein E-deficient mice. This could be a novel pathway to target in the design of drugs to limit AAA progression.


Asunto(s)
Aorta Abdominal/efectos de los fármacos , Aorta Abdominal/patología , Apolipoproteínas E/deficiencia , Factor XII/antagonistas & inhibidores , Proteína ADAM17/metabolismo , Angiotensina II/metabolismo , Angiotensina II/farmacología , Animales , Aneurisma de la Aorta Abdominal/metabolismo , Modelos Animales de Enfermedad , Factor XII/metabolismo , Ratones
8.
J Cardiovasc Pharmacol ; 73(5): 316-325, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30855407

RESUMEN

Chuanxiong rhizome has been widely used for the treatment of cerebral vascular disease in traditional Chinese medicine. The integrity of blood-brain barrier (BBB) is closely linked to the cerebral vascular disease. The protective effects of ligustilide, the major bioactive component in Chuanxiong rhizome, on cerebral blood vessels have been reported previously, but its effects and potential mechanism on BBB have not been entirely clarified. In the current work, the effects of ligustilide on BBB permeability and the underlying molecular mechanisms had been investigated using the model of BBB established by coculturing astrocytes and brain microvascular endothelial cells isolated from the rat brain. The ischemia-damaged model of BBB has been established with oxygen and glucose deprivation (OGD). Our results indicated that OGD significantly increased the permeability in the coculture BBB model. This OGD-induced increase in permeability could suppress by ligustilide in a concentration-dependent manner. Also, ligustilide promoted both gene and protein expression of tight junction proteins. Ligustilide suppressed the upregulation of HIF-1α, vascular endothelial growth factor, and AQP-4 in the BBB model induced by OGD. Collectively, all results have demonstrated that ligustilide is capable of reducing the permeability of BBB in vitro model induced by OGD through HIF-1α/vascular endothelial growth factor pathway and AQP-4, which provide a new target for the clinical application of ligustilide on BBB after stroke in future.


Asunto(s)
4-Butirolactona/análogos & derivados , Astrocitos/efectos de los fármacos , Barrera Hematoencefálica/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Glucosa/deficiencia , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , 4-Butirolactona/farmacología , Animales , Acuaporina 4/metabolismo , Astrocitos/metabolismo , Astrocitos/patología , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Hipoxia de la Célula , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Masculino , Ratas Sprague-Dawley , Transducción de Señal , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Uniones Estrechas/patología , Proteína de la Zonula Occludens-1/genética , Proteína de la Zonula Occludens-1/metabolismo
9.
Molecules ; 24(6)2019 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-30897753

RESUMEN

The Naoxinqing (NXQ) tablet is a standardised proprietary herbal product containing an extract of persimmon leaves (Diospyros kaki) for the management of cardio- and cerebrovascular diseases. Although previous reports suggested that the efficacy of NXQ is at least partly mediated by its anti-oxidative property, the anti-oxidative effect of the major components of NXQ has not been studied systematically. For quality control purposes, only analytical methods limited to 3 marker analytes have been reported, the extent to which the other components affect efficacy has not been explored. In this study, we developed an ultra-performance liquid chromatography-tandem mass spectrometry (UPLC MS/MS) method for the identification of seven analytes (kaempferol-3-O-glucoside (astragalin), quercetin-3-O-galactoside (hypericin), quercetin-3-O-glucoside (isoquercitin), kaempferol, 3,4-dihydroxybenzoic acid (protocatechuic acid), and furan-2-carboxylic acid (pyromucic acid) and quercetin) in the NXQ. This is the first method reported and validated for the quantification of the seven major secondary metabolites in NXQ. The results for the quantified analytes were then compared in 15 different batches of NXQ. The variation observed in the seven components highlights the need to quantify key bioactive components to ensure product consistency. Radical scavenging activity and abundance was used to rank the analytes. The anti-oxidative effects of NXQ were examined using cultured human vascular endothelial cells (EA.hy926). Corrected 2,2-di(4-tert-octylphenyl)-1-picrylhydrazyl (DPPH) activity results revealed that quercetin and kaempferol have the strongest anti-oxidant capacity in the extract. Both quercetin and kaempferol significantly inhibited the hydrogen peroxide (H2O2)-induced EA.hy926 cell injury and intracellular reactive oxygen species (ROS) generation. In conclusion, we established and validated an UPLC-MS/MC method for the analysis of major bioactive components in the NXQ and demonstrated that its anti-oxidative property may play a critical role in cerebrovascular protection.


Asunto(s)
Diospyros/química , Medicamentos Herbarios Chinos/química , Extractos Vegetales/química , Hojas de la Planta/química , Antracenos , Línea Celular , Cromatografía Líquida de Alta Presión , Humanos , Hidroxibenzoatos/química , Quempferoles/química , Perileno/análogos & derivados , Perileno/química , Quercetina/análogos & derivados , Quercetina/química , Especies Reactivas de Oxígeno , Espectrometría de Masas en Tándem
10.
Arterioscler Thromb Vasc Biol ; 37(3): 553-566, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28062506

RESUMEN

OBJECTIVE: Sclerostin (SOST) has been identified as an important regulator of bone formation; however, it has not been previously implicated in arterial disease. The aim of this study was to assess the role of SOST in aortic aneurysm (AA) and atherosclerosis using human samples, a mouse model, and in vitro investigations. APPROACH AND RESULTS: SOST protein was downregulated in human and mouse AA samples compared with controls. Transgenic introduction of human SOST in apolipoprotein E-deficient (ApoE-/-) mice (SOSTTg .ApoE-/-) and administration of recombinant mouse Sost inhibited angiotensin II-induced AA and atherosclerosis. Serum concentrations of several proinflammatory cytokines were significantly reduced in SOSTTg .ApoE-/- mice. Compared with controls, the aortas of mice receiving recombinant mouse Sost and SOSTTg .ApoE-/- mice showed reduced matrix degradation, reduced elastin breaks, and preserved collagen. Decreased inflammatory cell infiltration and a reduction in the expression of wingless-type mouse mammary virus integration site/ß-catenin responsive genes, including matrix metalloproteinase-9, osteoprotegerin, and osteopontin, were observed in the aortas of SOSTTg .ApoE-/- mice. SOST expression was downregulated and the wingless-type mouse mammary virus integration site/ß-catenin pathway was activated in human AA samples. The cytosine-phosphate-guanine islands in the SOST gene promoter showed significantly higher methylation in human AA samples compared with controls. Incubation of vascular smooth muscle cells with the demethylating agent 5-azacytidine resulted in upregulation of SOST, suggesting that SOST is epigenetically regulated. CONCLUSIONS: This study identifies that SOST is expressed in the aorta and downregulated in human AA possibly because of epigenetic silencing. Upregulating SOST inhibits AA and atherosclerosis development, with potential important implications for treating these vascular diseases.


Asunto(s)
Angiotensina II , Aneurisma de la Aorta/prevención & control , Aterosclerosis/prevención & control , Proteínas Morfogenéticas Óseas/metabolismo , Glicoproteínas/administración & dosificación , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales , Anciano , Anciano de 80 o más Años , Animales , Aorta Abdominal/efectos de los fármacos , Aorta Abdominal/metabolismo , Aorta Abdominal/patología , Aorta Torácica/efectos de los fármacos , Aorta Torácica/metabolismo , Aorta Torácica/patología , Aneurisma de la Aorta/inducido químicamente , Aneurisma de la Aorta/genética , Aneurisma de la Aorta/metabolismo , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/inducido químicamente , Aterosclerosis/genética , Aterosclerosis/metabolismo , Proteínas Morfogenéticas Óseas/genética , Células Cultivadas , Citocinas/metabolismo , Epigénesis Genética/efectos de los fármacos , Matriz Extracelular/metabolismo , Femenino , Marcadores Genéticos/genética , Predisposición Genética a la Enfermedad , Humanos , Péptidos y Proteínas de Señalización Intercelular , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Fenotipo , Remodelación Vascular/efectos de los fármacos
11.
Chem Pharm Bull (Tokyo) ; 66(10): 983-991, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30270244

RESUMEN

A novel strategy for screening active components in traditional Chinese medicines (TCM) using living cells and HPLC and GC analysis are proposed. The hypothesis is that when cells are incubated with the extract of Tongqiao Huoxue Decoction (TQHXD), a famous ancient prescription in TCM, the potential active components in the TQHXD should selectively combine with the cells, and the cell-combining components would be detectable in the extract of denatured cells. The identities of the cell-combining components could be determined by HPLC and GC analysis. Using the proposed approach, two characteristic active ingredients binding to the membrane of the PC12 cells are indicated. In the fingerprint of HPLC, there are two characteristic peaks. One active ingredient with its retention time was at around 70 min had been identified as muscone by HPLC, GC, which came from Moschus herb, the other active ingredient may come from the Allium fistulosum, its structure needs further research. Also, the protective effect of muscone on PC12 cells induced by Oxygen and glucose deprivation (OGD) had been explored. These results show that the pretreatment with muscone on PC12 cells observably increased cell viability, reduced the release of lactate dehydrogenase (LDH) and cell apoptosis. Combined with the pharmacodynamic study of muscone on neuroprotective effect, it could be identified as one of the effective components in TQHXD.


Asunto(s)
Allium/química , Medicamentos Herbarios Chinos/farmacología , Neuronas/efectos de los fármacos , Extractos Vegetales/farmacología , Animales , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cromatografía Líquida de Alta Presión , Relación Dosis-Respuesta a Droga , Medicamentos Herbarios Chinos/análisis , Medicina Tradicional China , Células PC12 , Extractos Vegetales/análisis , Ratas , Relación Estructura-Actividad
12.
Clin Sci (Lond) ; 131(12): 1261-1281, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28364044

RESUMEN

Abdominal aortic aneurysm (AAA) is a common age-related vascular disease characterized by progressive weakening and dilatation of the aortic wall. Thrombospondin-1 (TSP-1; gene Thbs1) is a member of the matricellular protein family important in the control of extracellular matrix (ECM) remodelling. In the present study, the association of serum TSP-1 concentration with AAA progression was assessed in 276 men that underwent repeated ultrasound for a median 5.5 years. AAA growth was negatively correlated with serum TSP-1 concentration (Spearman's rho -0.129, P=0.033). Men with TSP-1 in the highest quartile had a reduced likelihood of AAA growth greater than median during follow-up (OR: 0.40; 95% confidence interval (CI): 0.19-0.84, P=0.016, adjusted for other risk factors). Immunohistochemical staining for TSP-1 was reduced in AAA body tissues compared with the relatively normal AAA neck. To further assess the role of TSP-1 in AAA initiation and progression, combined TSP-1 and apolipoprotein deficient (Thbs1-/-ApoE-/-, n=20) and control mice (ApoE-/-, n=20) were infused subcutaneously with angiotensin II (AngII) for 28 days. Following AngII infusion, Thbs1-/- ApoE-/- mice had larger AAAs by ultrasound (P=0.024) and ex vivo morphometry measurement (P=0.006). The Thbs1-/-ApoE-/- mice also showed increased elastin filament degradation along with elevated systemic levels and aortic expression of matrix metalloproteinase (MMP)-9. Suprarenal aortic segments and vascular smooth muscle cells (VSMCs) isolated from Thbs1-/-ApoE-/- mice showed reduced collagen 3A1 gene expression. Furthermore, Thbs1-/-ApoE-/- mice had reduced aortic expression of low-density lipoprotein (LDL) receptor-related protein 1. Collectively, findings from the present study suggest that TSP-1 deficiency promotes maladaptive remodelling of the ECM leading to accelerated AAA progression.


Asunto(s)
Angiotensina II , Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/metabolismo , Trombospondina 1/sangre , Trombospondina 1/deficiencia , Animales , Aorta Abdominal/diagnóstico por imagen , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/sangre , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/prevención & control , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Biomarcadores/sangre , Células Cultivadas , Colágeno Tipo III/genética , Colágeno Tipo III/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Elastina/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Masculino , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Noqueados , Oportunidad Relativa , Fenotipo , Proteolisis , Receptores de LDL/genética , Receptores de LDL/metabolismo , Factores de Riesgo , Trombospondina 1/genética , Factores de Tiempo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Ultrasonografía , Remodelación Vascular
13.
Int J Mol Sci ; 18(1)2017 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-28067784

RESUMEN

Sailuotong (SLT) is a standardised three-herb formulation consisting of Panax ginseng, Ginkgo biloba, and Crocus sativus designed for the management of vascular dementia. While the latest clinical trials have demonstrated beneficial effects of SLT in vascular dementia, the underlying cellular mechanisms have not been fully explored. The aim of this study was to assess the ability and mechanisms of SLT to act against hydrogen peroxide (H2O2)-induced oxidative damage in cultured human vascular endothelial cells (EAhy926). SLT (1-50 µg/mL) significantly suppressed the H2O2-induced cell death and abolished the H2O2-induced reactive oxygen species (ROS) generation in a concentration-dependent manner. Similarly, H2O2 (0.5 mM; 24 h) caused a ~2-fold increase in lactate dehydrogenase (LDH) release from the EA.hy926 cells which were significantly suppressed by SLT (1-50 µg/mL) in a concentration-dependent manner. Incubation of SLT (50 µg/mL) increased superoxide dismutase (SOD) activity and suppressed the H2O2-enhanced Bax/Bcl-2 ratio and cleaved caspase-3 expression. In conclusion, our results suggest that SLT protects EA.hy916 cells against H2O2-mediated injury via direct reduction of intracellular ROS generation and an increase in SOD activity. These protective effects are closely associated with the inhibition of the apoptotic death cascade via the suppression of caspase-3 activation and reduction of Bax/Bcl-2 ratio, thereby indicating a potential mechanism of action for the clinical effects observed.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Células Endoteliales/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Western Blotting , Caspasa 3/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células Endoteliales/metabolismo , Humanos , L-Lactato Deshidrogenasa/metabolismo , Oxidantes/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo , Proteína X Asociada a bcl-2/metabolismo
14.
Arterioscler Thromb Vasc Biol ; 35(2): 389-98, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25524772

RESUMEN

OBJECTIVE: Interaction of the activating sequence in thrombospondin-1 (TSP-1) with the conserved sequence (leucine-serine-lysine-leucine [LSKL]) in the latency-associated peptide region of latent transforming growth factor (TGF)-ß complex is important in regulating TGF-ß1 activity. We aimed to assess the effect of blocking peptide LSKL on the progression of pre-established abdominal aortic aneurysm in angiotensin II-infused apolipoprotein E-deficient (ApoE(-/-)) mice. APPROACH AND RESULTS: Abdominal aortic aneurysm was established in 3-month-old male ApoE(-/-) mice with subcutaneous infusion of angiotensin II for 28 days. After this, mice received LSKL peptide or control SLLK (serine-leucine-leucine-lysine) peptide (4 mg/kg) via daily intraperitoneal injection for an additional 2 weeks. Administration of LSKL peptide promoted larger suprarenal aortic diameter, as determined by ultrasound and morphometric analysis, and stimulated more severe atherosclerosis within the aortic arch. In addition, mice receiving LSKL peptide exhibited elevated circulating proinflammatory cytokine levels and greater inflammatory cells within the suprarenal aorta compared with controls. Mice receiving LSKL peptide showed low plasma TGF-ß1 activity and low levels of aortic tissue phosphorylated to total Smad2/3. Aortic gene expression of TGF-ß receptor 1 (TGFBRI) and receptor 2 (TGFBRII), but not TGF-ß1 and thrombospondin-1, were lower in mice receiving LSKL peptide than controls. LSKL peptide administration was associated with greater aortic elastin fragmentation and lower expression and activity of the TGF-ß1-target gene lysyl oxidase like 1 (LOXL1). CONCLUSIONS: Attenuation of thrombospondin-1-directed activation of TGF-ß1 promotes abdominal aortic aneurysm and atherosclerosis progression in the angiotensin II-infused ApoE(-/-) mouse model.


Asunto(s)
Angiotensina II , Aorta/efectos de los fármacos , Aneurisma de la Aorta Abdominal/inducido químicamente , Apolipoproteínas E/deficiencia , Aterosclerosis/inducido químicamente , Péptidos/toxicidad , Trombospondina 1/antagonistas & inhibidores , Aminoácido Oxidorreductasas/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Aneurisma de la Aorta Abdominal/sangre , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/patología , Apolipoproteínas E/genética , Aterosclerosis/sangre , Aterosclerosis/genética , Aterosclerosis/patología , Citocinas/sangre , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Elastina/metabolismo , Mediadores de Inflamación/sangre , Inyecciones Intraperitoneales , Masculino , Ratones Noqueados , Péptidos/administración & dosificación , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Trombospondina 1/metabolismo , Factores de Tiempo , Factor de Crecimiento Transformador beta1/sangre
15.
Heart Fail Rev ; 19(2): 187-98, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23225134

RESUMEN

Sympathetic nerve activity has been reported to be increased in both humans and animals with chronic heart failure. One of the mechanisms believed to be responsible for this phenomenon is increased systemic and cerebral angiotensin II signaling. Plasma angiotensin II is increased in humans and animals with chronic heart failure. The increase in angiotensin II signaling enhances sympathetic nerve activity through actions on both central and peripheral sites during chronic heart failure. Angiotensin II signaling is enhanced in different brain sites such as the paraventricular nucleus, the rostral ventrolateral medulla and the area postrema. Blocking angiotensin II type 1 receptors decreases sympathetic nerve activity and cardiac sympathetic afferent reflex when therapy is administered to the paraventricular nucleus. Injection of an angiotensin receptor blocker into the area postrema activates the sympathoinhibitory baroreflex. In peripheral regions, angiotensin II elevates both norepinephrine release and synthesis and inhibits norepinephrine uptake at nerve endings, which may contribute to the increase in sympathetic nerve activity seen in chronic heart failure. Increased circulating angiotensin II during chronic heart failure may enhance the sympathoexcitatory chemoreflex and inhibit the sympathoinhibitory baroreflex. In addition, increased circulating angiotensin II can directly act on the central nervous system via the subfornical organ and the area postrema to increase sympathetic outflow. Inhibition of angiotensin II formation and its type 1 receptor has been shown to have beneficial effects in chronic heart failure patients.


Asunto(s)
Angiotensina II/sangre , Insuficiencia Cardíaca/fisiopatología , Corazón/fisiopatología , Sistema Nervioso Simpático/fisiopatología , Enfermedad Crónica , Humanos
16.
Clin Sci (Lond) ; 127(2): 123-34, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24476071

RESUMEN

Aliskiren is a direct renin inhibitor developed to treat hypertension. Several clinical studies have suggested that aliskiren has beneficial effects on cardiovascular diseases beyond its antihypertensive effect. In the present study, we examined whether aliskiren limits the progression of AAA (abdominal aortic aneurysm), VH (ventricular hypertrophy) and atherosclerosis in an AngII (angiotensin II)-infused mouse model. ApoE-/- (apolipoprotein-E-deficient) mice were infused subcutaneously with AngII (1000 ng/kg of body weight per day; 4 weeks) to induce AAA and VH. At the completion of the AngII infusion, mice were randomly allocated to three groups to receive vehicle control, low-dose aliskiren (10 mg/kg of body weight per day) or high-dose aliskiren (50 mg/kg of body weight per day) for 4 weeks. Suprarenal aortic diameter assessed by ultrasound was significantly smaller in mice administered aliskiren at days 42 and 56. Aliskiren also significantly reduced the normalized heart weight, ventricular myocyte cell width and aortic arch atherosclerosis. Aliskiren lowered PRR (pro-renin receptor) expression and MAPK (mitogen-activated protein kinase) activity in the suprarenal aorta and heart. Aortic infiltration of T-lymphocytes and macrophages was reduced by aliskiren. In conclusion, aliskiren limits the progression of AAA, VH and atherosclerosis in an AngII-infused mouse model.


Asunto(s)
Amidas/farmacología , Aneurisma de la Aorta Abdominal/tratamiento farmacológico , Apolipoproteínas E/metabolismo , Aterosclerosis/tratamiento farmacológico , Fumaratos/farmacología , Hipertrofia/tratamiento farmacológico , Angiotensina II/metabolismo , Animales , Aorta Abdominal/efectos de los fármacos , Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/metabolismo , Apolipoproteínas E/deficiencia , Aterosclerosis/metabolismo , Presión Sanguínea/efectos de los fármacos , Modelos Animales de Enfermedad , Hipertensión/metabolismo , Hipertrofia/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
17.
Biomed Pharmacother ; 172: 116269, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38367549

RESUMEN

AGS-30, a new andrographolide derivative, showed significant anticancer and anti-angiogenic characteristics. However, its role in controlling macrophage polarization and tumor immune response is unknown. Thus, the main goals of this study are to investigate how AGS-30 regulates macrophage polarization and how it suppresses breast cancer metastasis. AGS-30 inhibited IL-4 and IL-13-induced RAW 264.7 and THP-1 macrophages into M2-like phenotype. However, AGS-30 did not affect the LPS and IFN-γ-induced polarization of M1-like macrophages. AGS-30 reduced the mRNA expressions of CD206, Arg-1, Fizz-1, Ym-1, VEGF, IL-10, MMP2, and MMP9 in M2-like macrophages in a concentration-dependent manner. In contrast, andrographolide treatment at 5 µM did not affect M1-like and M2-like macrophage polarization. The conditioned medium from M2-like macrophages increased 4T1 breast cancer cell migration and invasion, whereas AGS-30 inhibited these effects. In the 4T1 breast tumor xenograft mice, the tumor volume and weight were reduced without affecting body weight after receiving AGS-30. AGS-30 treatment also reduced lung and liver metastasis, with reduced STAT6, CD31, VEGF, and Ki67 protein expressions. Moreover, the tumors had considerably fewer M2-like macrophages and Arg-1 expression, but the proportion of M1-like macrophages and iNOS expression increased after AGS-30 treatment. Same results were found in the tail vein metastasis model. In conclusion, this study shows that AGS-30 inhibits breast cancer growth and metastasis, probably through inhibiting M2-like macrophage polarization. Our findings suggest that AGS-30 may be a potential immunotherapeutic alternative for metastatic breast cancer.


Asunto(s)
Neoplasias de la Mama , Diterpenos , Animales , Femenino , Humanos , Ratones , Neoplasias de la Mama/tratamiento farmacológico , Medios de Cultivo Condicionados , Diterpenos/farmacología , Neoplasias Mamarias Animales/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular
18.
Biomed Pharmacother ; 176: 116901, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38878683

RESUMEN

BACKGROUND: Amauroderma rugosum (AR) is a medicinal mushroom commonly used to treat inflammation, gastric disorders, epilepsy, and cancers due to its remarkable anti-inflammatory and anti-oxidative properties. This study was designed to evaluate the pharmacological effects of AR and its underlying mechanism of action against ulcerative colitis (UC) in vitro and in vivo. METHODS: A UC mouse model was established by administration of dextran sulfate sodium (DSS). AR extract was administered intragastrically to mice for 7 days. At the end of the experiment, histopathology, macrophage phenotype, oxidative stress, and inflammatory status were examined in vivo. Furthermore, RAW 264.7, THP-1, and Caco-2 cells were used to elucidate the mechanism of action of AR in vitro. RESULTS: AR extract (0.5-2 mg/mL) significantly suppressed lipopolysaccharide (LPS) and interferon-gamma (IFN-γ)-induced M1 macrophage (pro-inflammatory) polarization in both RAW 264.7 and THP-1 cells. LPS-induced pro-inflammatory mediators (nitric oxide, TNF-α, IL-1ß, MCP-1, and IL-6) were reduced by AR extract in a concentration-dependent manner. Similarly, AR extract downregulated MAPK signaling activity in LPS-stimulated RAW 264.7 cells. AR extract elicited a concentration-dependent increase in the mRNA expression of M2 (anti-inflammatory) phenotype markers (CD206, Arg-1, Fizz-1, and Ym-1) in RAW 264.7 cells. Moreover, AR extract suppressed DSS-induced ROS generation and mitochondrial dysfunction in Caco-2 cells. The in vivo experiment revealed that AR extract (200 mg/kg) increased colon length compared to the DSS-treated group. In addition, disease activity index, spleen ratio, body weight, oxidative stress, and colonic inflammation were markedly improved by AR treatment in DSS-induced UC mice. Finally, AR suppressed M1 and promoted M2 macrophage polarization in UC mice. CONCLUSION: The AR extract protected against DSS-induced UC by regulating macrophage polarization and suppressing oxidative stress. These valuable findings suggest that adequate intake of AR can prevent and/or treat UC.


Asunto(s)
Colitis Ulcerosa , Sulfato de Dextran , Macrófagos , Estrés Oxidativo , Animales , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/patología , Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/prevención & control , Estrés Oxidativo/efectos de los fármacos , Ratones , Humanos , Células CACO-2 , Células RAW 264.7 , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Células THP-1 , Antiinflamatorios/farmacología , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Citocinas/metabolismo , Mediadores de Inflamación/metabolismo
19.
Int J Biol Macromol ; 271(Pt 2): 132533, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38777026

RESUMEN

Amauroderma rugosum (AR), also known as "Blood Lingzhi" in Chinese, is a basidiomycete belonging to the Ganodermataceae family. Four polysaccharide fractions were systematically isolated and purified from AR. Subsequently, their compositions were examined and analyzed via high-performance gel permeation chromatography (HPGPC), analysis of the monosaccharide composition, Fourier-transform infrared spectroscopy (FT-IR), and 1H nuclear magnetic resonance (NMR). The zebrafish model was then used to screen for proangiogenic activities of polysaccharides by inducing vascular insufficiency with VEGF receptor tyrosine kinase inhibitor II (VRI). The third fraction of AR polysaccharides (PAR-3) demonstrated the most pronounced proangiogenic effects, effectively ameliorating VRI-induced intersegmental vessel deficiency in zebrafish. Concurrently, the mRNA expression levels of vascular endothelial growth factor (VEGF)-A and VEGF receptors were upregulated by PAR-3. Moreover, the proliferation, migration, invasion, and tube formation of human umbilical vein endothelial cells (HUVECs) were also stimulated by PAR-3, consistently demonstrating that PAR-3 possesses favorable proangiogenic properties. The activation of the Akt, ERK1/2, p38 MAPK, and FAK was most likely the underlying mechanism. In conclusion, this study establishes that PAR-3 isolated from Amauroderma rugosum exhibits potential as a bioresource for promoting angiogenesis.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana , Pez Cebra , Animales , Humanos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Polisacáridos/farmacología , Polisacáridos/química , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Neovascularización Fisiológica/efectos de los fármacos , Inductores de la Angiogénesis/farmacología , Inductores de la Angiogénesis/química , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Basidiomycota/química , Polisacáridos Fúngicos/farmacología , Polisacáridos Fúngicos/química
20.
Am J Pathol ; 181(2): 706-18, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22698985

RESUMEN

There are currently no acceptable treatments to limit progression of abdominal aortic aneurysm (AAA). Increased serum concentrations of high-density lipoprotein (HDL) are associated with reduced risk of developing an AAA. The present study aimed to assess the effects of fenofibrate on aortic dilatation in a mouse model of AAA. Male low-density lipoprotein receptor-deficient (Ldlr(-/-)) mice were maintained on a high-fat diet for 3 weeks followed by 6 weeks of oral administration of vehicle or fenofibrate. From 14 to 18 weeks of age, all mice were infused with angiotensin II (AngII). At 18 weeks of age, blood and aortas were collected for assessment of serum lipoproteins, aortic pathology, aortic Akt1 and endothelial nitric oxide synthase (eNOS) activities, immune cell infiltration, eNOS and inducible NOS (iNOS) expression, sphingosine 1 phosphate (S1P) receptor status, and apoptosis. Mice receiving fenofibrate had reduced suprarenal aortic diameter, reduced aortic arch Sudan IV staining, higher serum HDL levels, increased serum S1P concentrations, and increased aortic Akt1 and eNOS activities compared with control mice. Macrophages, T lymphocytes, and apoptotic cells were less evident and eNOS, iNOS, and S1P receptors 1 and 3 were up-regulated in aortas from mice receiving fenofibrate. The present findings suggest that fenofibrate antagonizes AngII-induced AAA and atherosclerosis by up-regulating serum HDL and S1P levels, with associated activation of NO-producing enzymes and reduction of aortic inflammation.


Asunto(s)
Aneurisma de la Aorta Abdominal/sangre , Aneurisma de la Aorta Abdominal/patología , Progresión de la Enfermedad , Fenofibrato/uso terapéutico , Lipoproteínas HDL/sangre , Lisofosfolípidos/sangre , Esfingosina/análogos & derivados , Angiotensina II , Animales , Aorta/efectos de los fármacos , Aorta/enzimología , Aorta/patología , Aorta Torácica/efectos de los fármacos , Aorta Torácica/patología , Aneurisma de la Aorta Abdominal/complicaciones , Aneurisma de la Aorta Abdominal/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Compuestos Azo/metabolismo , Dilatación Patológica/sangre , Dilatación Patológica/complicaciones , Dilatación Patológica/tratamiento farmacológico , Dilatación Patológica/patología , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Células Endoteliales/patología , Fenofibrato/farmacología , Inflamación/sangre , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Inflamación/patología , Riñón/irrigación sanguínea , Riñón/efectos de los fármacos , Riñón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/sangre , Resultado del Tratamiento , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA