Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Postgrad Med J ; 100(1183): 297-304, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38263934

RESUMEN

BACKGROUND: Dysmobility syndrome based on osteoporosis (ODS) is a disease characterized by low bone mass and low muscle mass. Its features are high fracture and high fall risk. Falls and fractures are the most important factors affecting the quality of life and lifespan of ODS. However, there is no serum marker for the evaluation of ODS patients.Our previous studies have shown that the expression of circulating miRNA is stable and is a good marker for disease diagnosis. Therefore, this study aims to explore potential serum markers of ODS. METHODS: A total of 78 subjects were included in this study. The data including appendicular skeletal muscle mass index, bone mineral density, bone metabolism markers, and other relevant information were collected for analysis. Real-time quantitative polymerase chain reaction was used to detect 19 miRNAs associated with muscle mass reduction. The correlation of quantitative data was analyzed by Pearson. The receiver operating characteristic curve was used to evaluate the performance of miRNA as a biomarker. RESULTS: In this study, we found that the muscle mass and strength of patients with ODS are significantly reduced and are negatively correlated with the risk of fracture. The hsa-miR-499a-5p is specifically downregulated in ODS, and is positively correlated with muscle mass and strength, and negatively correlated with the risk of fracture. Compared with muscle mass and strength, hsa-miR-499a-5p has better sensitivity and specificity as a diagnostic marker. CONCLUSION: hsa-miR-499a-5p is a potential serum biomarker for assessing muscle function and predicting fall or fracture risk in the ODS population.


Asunto(s)
Biomarcadores , MicroARNs , Osteoporosis , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Biomarcadores/sangre , Densidad Ósea , Fracturas Óseas/etiología , Fracturas Óseas/sangre , MicroARNs/sangre , Músculo Esquelético , Osteoporosis/sangre , Osteoporosis/diagnóstico , Síndrome
2.
J Neurosci ; 2022 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-35985835

RESUMEN

Traumatic brain injury (TBI) triggers a plethora of inflammatory events in the brain that aggravate secondary injury and impede tissue repair. Resident microglia (Mi) and blood-borne infiltrating macrophages (MΦ) are major players of inflammatory responses in the post-TBI brain and possess high functional heterogeneity. However, the plasticity of these cells has yet to be exploited to develop therapies that can mitigate brain inflammation and improve the outcome after TBI. This study investigated the transcription factor STAT1 as a key determinant of proinflammatory Mi/MΦ responses and aimed to develop STAT1 as a novel therapeutic target for TBI using a controlled cortical impact model of TBI on adult male mice. TBI induced robust upregulation of STAT1 in the brain at the subacute injury stage, which occurred primarily in Mi/MΦ. Intraperitoneal administration of fludarabine, a selective STAT1 inhibitor, markedly alleviated proinflammatory Mi/MΦ responses and brain inflammation burden after TBI. Such phenotype-modulating effects of fludarabine on post-TBI Mi/MΦ were reproduced by tamoxifen-induced, selective knockout of STAT1 in Mi/MΦ (STAT1 mKO). By propelling Mi/MΦ away from a detrimental proinflammatory phenotype, STAT1 mKO was sufficient to reduce long-term neurological deficits and brain lesion size after TBI. Importantly, short-term fludarabine treatment after TBI elicited long-lasting improvement of TBI outcomes, but this effect was lost on STAT1 mKO mice. Together, our study provided the first line of evidence that STAT1 causatively determines the proinflammatory phenotype of brain Mi/MΦ after TBI. We also showed promising preclinical data supporting the use of fludarabine as a novel immunomodulating therapy to TBI.SIGNIFICANCE STATEMENTThe functional phenotype of microglia and macrophages (Mi/MΦ) critically influences brain inflammation and the outcome after traumatic brain injury (TBI); however, no therapies have been developed to modulate Mi/MΦ functions to treat TBI. Here we report for the first time that the transcription factor STAT1 is a key mediator of proinflammatory Mi/MΦ responses in the post-TBI brain, the specific deletion of which ameliorates neuroinflammation and improves long-term functional recovery after TBI. We also show excellent efficacy of a selective STAT1 inhibitor fludarabine against TBI-induced functional deficits and brain injury using a mouse model, presenting STAT1 as a promising therapeutic target for TBI.

3.
J Neuroinflammation ; 20(1): 178, 2023 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-37516843

RESUMEN

BACKGROUND: Brain microglia and macrophages (Mi/MΦ) can shift to a harmful or advantageous phenotype following an ischemic stroke. Identification of key molecules that regulate the transformation of resting Mi/MΦ could aid in the development of innovative therapies for ischemic stroke. The transcription factor signal transducer and activator of transduction 1 (STAT1) has been found to contribute to acute neuronal death (in the first 24 h) following ischemic stroke, but its effects on Mi/MΦ and influence on long-term stroke outcomes have yet to be determined. METHODS: We generated mice with tamoxifen-induced, Mi/MΦ-specific knockout (mKO) of STAT1 driven by Cx3cr1CreER. Expression of STAT1 was examined in the brain by flow cytometry and RNA sequencing after ischemic stroke induced by transient middle cerebral artery occlusion (MCAO). The impact of STAT1 mKO on neuronal cell death, Mi/MΦ phenotype, and brain inflammation profiles were examined 3-5 days after MCAO. Neurological deficits and the integrity of gray and white matter were assessed for 5 weeks after MCAO by various neurobehavioral tests and immunohistochemistry. RESULTS: STAT1 was activated in Mi/MΦ at the subacute stage (3 days) after MCAO. Selective deletion of STAT1 in Mi/MΦ did not alter neuronal cell death or infarct size at 24 h after MCAO, but attenuated Mi/MΦ release of high mobility group box 1 and increased arginase 1-producing Mi/MΦ 3d after MCAO, suggesting boosted inflammation-resolving responses of Mi/MΦ. As a result, STAT1 mKO mice had mitigated brain inflammation at the subacute stage after MCAO and less white matter injury in the long term. Importantly, STAT1 mKO was sufficient to improve functional recovery for at least 5 weeks after MCAO in both male and female mice. CONCLUSIONS: Mi/MΦ-targeted STAT1 KO does not provide immediate neuroprotection but augments inflammation-resolving actions of Mi/MΦ, thereby facilitating long-term functional recovery after stroke. STAT1 is, therefore, a promising therapeutic target to harness beneficial Mi/MΦ responses and improve long-term outcomes after ischemic stroke.


Asunto(s)
Encefalitis , Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Animales , Femenino , Masculino , Ratones , Inflamación , Macrófagos , Microglía
4.
Osteoporos Int ; 34(4): 627-639, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36380163

RESUMEN

PURPOSE: The aim is to evaluate the effect of ß-carotene for osteoporosis and provide quantitative evidence. METHOD: PubMed, Embase, Web of Science, and Cochrane Library were searched for eligible studies. Fifteen studies were included. Random-effect model was applied to pool the odds ratio (OR). The risk of osteoporosis and fracture were compared between low ß-carotene intake group and high ß-carotene intake group. RESULT: The intake of ß-carotene was unassociated with the overall risk of osteoporosis [OR = 0.733, 95% Cl (0.528, 1.018), p = 0.064]. Subgroup analysis showed that the intake of ß-carotene was negatively associated with the risk of osteoporosis in both male subgroup [OR = 0.7, 95% Cl (0.549, 0.893), I2 = 40.40%, p = 0.004] and female subgroup [OR = 0.684, 95% Cl (0.487, 0.960), I2 = 86.40%, p = 0.028]. There was also a negative association between ß-carotene intake and osteoporosis in Asia subgroup [OR = 0.512, 95% Cl (0.403, 0.650), I2 = 0.00%, p = 0], whereas no association was observed in Western subgroup [OR = 1.107, 95% Cl (0.908, 1.350), I2 = 2.30%, p = 0.314]. In addition, random-effect model was adopted to pool the standard mean difference (SMD), and the results showed that ß-carotene intake was positively associated with overall bone mineral density (BMD) [SMD = - 0.213, 95% Cl (- 0.391, - 0.034), I2 = 87.30%, p = 0.019]. Subgroup analysis showed that ß-carotene intake was positively associated with BMD in Asian participants [SMD = - 0.394, 95% Cl (- 0.461, - 0.328), I2 = 0, p = 0], while unassociated in Western participants [SMD = - 0.047, 95% Cl (- 0.314, 0.219), I2 = 78.9%, p = 0.727]. CONCLUSION: ß-carotene may improve BMD and reduce the risk of osteoporosis and fracture. However, these effects could vary by gender and race and need to be further validated by longitudinal studies.


Asunto(s)
Fracturas Óseas , Osteoporosis , Masculino , Humanos , Femenino , beta Caroteno/farmacología , beta Caroteno/uso terapéutico , Densidad Ósea , Asia
5.
Qual Life Res ; 32(6): 1551-1565, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36383282

RESUMEN

PURPOSE: Postmenopausal osteoporosis has become a global trend, which seriously affects women's quality of life. However, the differences remain unclear in health-related quality of life (HRQoL) among postmenopausal women with normal bone mineral density, osteoporosis, and osteoporotic fractures. The aim of this study was to assess health-related quality of life in women with three different bone states. METHODS: Databases of PubMed, Embase, Cochrane, and Web of Science were based on the search terms, and the search time was set from the inception of each database to January 2022. A study was included if the researchers used a validated quality of life questionnaire to investigate the quality of life of postmenopausal women with osteoporosis or osteoporotic fractures. The random-effect model was used for meta-analysis, and the mean difference with a 95% confidence interval (95%CI) was calculated. RESULTS: Thirteen studies that met the inclusion criteria were systematically reviewed, involving 2897 postmenopausal women, and 12 of them were included in the meta-analysis. Postmenopausal women with osteoporosis had worse overall HRQoL and different HRQoL dimensions compared with postmenopausal women with normal bone density. Compared with postmenopausal women with osteoporosis, postmenopausal women with osteoporotic fractures had worse overall HRQoL and individual dimensions of HRQoL, especially physical component summary (SMD = - 0.61, 95% CI, - 0.98 to - 0.24). Bone mineral density was positively associated with HRQoL, while fragility fracture severity was negatively associated with HRQoL. CONCLUSIONS: Postmenopausal osteoporosis and fragility fractures reduce HRQoL to varying degrees in women. More research should be done to reduce the incidence of the disease.


Asunto(s)
Osteoporosis Posmenopáusica , Osteoporosis , Fracturas Osteoporóticas , Femenino , Humanos , Calidad de Vida/psicología , Osteoporosis Posmenopáusica/complicaciones , Osteoporosis Posmenopáusica/rehabilitación , Posmenopausia , Osteoporosis/complicaciones , Densidad Ósea
6.
J Neuroinflammation ; 19(1): 201, 2022 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-35933343

RESUMEN

BACKGROUND: Histone deacetylases (HDACs) are believed to exacerbate traumatic brain injury (TBI) based on studies using pan-HDAC inhibitors. However, the HDAC isoform responsible for the detrimental effects and the cell types involved remain unknown, which may hinder the development of specific targeting strategies that boost therapeutic efficacy while minimizing side effects. Microglia are important mediators of post-TBI neuroinflammation and critically impact TBI outcome. HDAC3 was reported to be essential to the inflammatory program of in vitro cultured macrophages, but its role in microglia and in the post-TBI brain has not been investigated in vivo. METHODS: We generated HDAC3LoxP mice and crossed them with CX3CR1CreER mice, enabling in vivo conditional deletion of HDAC3. Microglia-specific HDAC3 knockout (HDAC3 miKO) was induced in CX3CR1CreER:HDAC3LoxP mice with 5 days of tamoxifen treatment followed by a 30-day development interval. The effects of HDAC3 miKO on microglial phenotype and neuroinflammation were examined 3-5 days after TBI induced by controlled cortical impact. Neurological deficits and the integrity of white matter were assessed for 6 weeks after TBI by neurobehavioral tests, immunohistochemistry, electron microscopy, and electrophysiology. RESULTS: HDAC3 miKO mice harbored specific deletion of HDAC3 in microglia but not in peripheral monocytes. HDAC3 miKO reduced the number of microglia by 26%, but did not alter the inflammation level in the homeostatic brain. After TBI, proinflammatory microglial responses and brain inflammation were markedly alleviated by HDAC3 miKO, whereas the infiltration of blood immune cells was unchanged, suggesting a primary effect of HDAC3 miKO on modulating microglial phenotype. Importantly, HDAC3 miKO was sufficient to facilitate functional recovery for 6 weeks after TBI. TBI-induced injury to axons and myelin was ameliorated, and signal conduction by white matter fiber tracts was significantly enhanced in HDAC3 miKO mice. CONCLUSION: Using a novel microglia-specific conditional knockout mouse model, we delineated for the first time the role of microglial HDAC3 after TBI in vivo. HDAC3 miKO not only reduced proinflammatory microglial responses, but also elicited long-lasting improvement of white matter integrity and functional recovery after TBI. Microglial HDAC3 is therefore a promising therapeutic target to improve long-term outcomes after TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Histona Desacetilasas , Sustancia Blanca , Animales , Lesiones Traumáticas del Encéfalo/metabolismo , Modelos Animales de Enfermedad , Histona Desacetilasas/metabolismo , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Sustancia Blanca/metabolismo
7.
J Immunol ; 204(6): 1486-1498, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-32034062

RESUMEN

Microglia play essential roles in neuroinflammatory responses after traumatic brain injury (TBI). Our previous studies showed that phenotypes of microglia, as well as infiltrating macrophages, altered at different stages after CNS injury, which was correlated to functional outcomes. IL-13 is an anti-inflammatory cytokine that has been reported to protect against demyelination and spinal cord injury through immunomodulation. The effects of IL-13 in microglia/macrophage-mediated immune responses after TBI remain unknown. In this study, we showed that intranasal administration of IL-13 in male C57BL/6J mice accelerated functional recovery in the controlled cortical impact model of TBI. IL-13 treatment increased the time to fall off in the Rotarod test, reduced the number of foot faults in the foot fault test, and improved the score in the wire hang test up to 28 d after TBI. Consistent with functional improvement, IL-13 reduced neuronal tissue loss and preserved white matter integrity 6 d after TBI. Furthermore, IL-13 ameliorated the elevation of proinflammatory factors and reduced the number of proinflammatory microglia/macrophages 6 d after TBI. Additionally, IL-13 enhanced microglia/macrophage phagocytosis of damaged neurons in the peri-lesion areas. In vitro studies confirmed that IL-13 treatment inhibited the production of proinflammatory cytokines in rat primary microglia in response to LPS or dead neuron stimulation and increased the ability of microglia to engulf fluorophore-labeled latex beads or dead neurons. Collectively, we demonstrated that IL-13 treatment improved neurologic outcomes after TBI through adjusting microglia/macrophage phenotypes and inhibiting inflammatory responses. IL-13 may represent a potential immunotherapy to promote long-term recovery from TBI.


Asunto(s)
Antiinflamatorios/administración & dosificación , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Encefalitis/tratamiento farmacológico , Interleucina-13/administración & dosificación , Recuperación de la Función/efectos de los fármacos , Administración Intranasal , Animales , Técnicas de Observación Conductual , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Encéfalo/patología , Encéfalo/fisiopatología , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/fisiopatología , Células Cultivadas , Citocinas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalitis/inmunología , Encefalitis/fisiopatología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Ratones , Microglía/efectos de los fármacos , Microglía/inmunología , Microglía/metabolismo , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología , Cultivo Primario de Células , Ratas , Recuperación de la Función/inmunología
8.
Acta Pharmacol Sin ; 43(4): 1072-1081, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34183756

RESUMEN

Jingyin granules, a marketed antiviral herbal medicine, have been recommended for treating H1N1 influenza A virus infection and Coronavirus disease 2019 (COVID-19) in China. To fight viral diseases in a more efficient way, Jingyin granules are frequently co-administered in clinical settings with a variety of therapeutic agents, including antiviral drugs, anti-inflammatory drugs, and other Western medicines. However, it is unclear whether Jingyin granules modulate the pharmacokinetics of Western drugs or trigger clinically significant herb-drug interactions. This study aims to assess the inhibitory potency of the herbal extract of Jingyin granules (HEJG) against human drug-metabolizing enzymes and to clarify whether HEJG can modulate the pharmacokinetic profiles of Western drug(s) in vivo. The results clearly demonstrated that HEJG dose-dependently inhibited human CES1A, CES2A, CYPs1A, 2A6, 2C8, 2C9, 2D6, and 2E1; this herbal medicine also time- and NADPH-dependently inhibited human CYP2C19 and CYP3A. In vivo tests showed that HEJG significantly increased the plasma exposure of lopinavir (a CYP3A-substrate drug) by 2.43-fold and strongly prolonged its half-life by 1.91-fold when HEJG (3 g/kg) was co-administered with lopinavir to rats. Further investigation revealed licochalcone A, licochalcone B, licochalcone C and echinatin in Radix Glycyrrhizae, as well as quercetin and kaempferol in Folium Llicis Purpureae, to be time-dependent CYP3A inhibitors. Collectively, our findings reveal that HEJG modulates the pharmacokinetics of CYP substrate-drug(s) by inactivating CYP3A, providing key information for both clinicians and patients to use herb-drug combinations for antiviral therapy in a scientific and reasonable way.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Subtipo H1N1 del Virus de la Influenza A , Animales , Antivirales/farmacología , Inhibidores del Citocromo P-450 CYP3A , Interacciones de Hierba-Droga , Humanos , Microsomas Hepáticos , Ratas
9.
J Lipid Res ; 62: 100091, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34118252

RESUMEN

Plasma LDL is produced from catabolism of VLDL and cleared from circulation mainly via the hepatic LDL receptor (LDLR). Proprotein convertase subtilisin/kexin type 9 (PCSK9) promotes LDLR degradation, increasing plasma LDL-C levels. Circulating PCSK9 is mainly secreted by the liver, whereas VLDL is exclusively secreted by hepatocytes. However, the mechanism regulating their secretion is not completely understood. Surfeit 4 (Surf4) is a cargo receptor localized in the ER membrane. It recruits cargos into coat protein complex II vesicles to facilitate their secretion. Here, we investigated the role of Surf4 in VLDL and PCSK9 secretion. We generated Surf4 liver-specific knockout mice and found that knockout of Surf4 did not affect PCSK9 secretion, whereas it significantly reduced plasma levels of cholesterol, triglyceride, and lipid-binding protein apolipoprotein B (apoB). In cultured human hepatocytes, Surf4 coimmunoprecipitated and colocalized with apolipoprotein B100, and Surf4 silencing reduced secretion of apolipoprotein B100. Furthermore, knockdown of Surf4 in LDLR knockout (Ldlr-/-) mice significantly reduced triglyceride secretion, plasma levels of apoB and non-HDL-C, and the development of atherosclerosis. However, Surf4 liver-specific knockout mice and Surf4 knockdown in Ldlr-/- mice displayed similar levels of liver lipids and plasma alanine aminotransferase activity as control mice, indicating that inhibition of Surf4 does not cause notable liver damage. Expression of stearoyl-CoA desaturase-1 was also reduced in the liver of these mice, suggesting a reduction in de novo lipogenesis. In summary, hepatic deficiency of Surf4 reduced VLDL secretion and the development of atherosclerosis but did not cause significant hepatic lipid accumulation or liver damage.


Asunto(s)
Aterosclerosis/metabolismo , Lipoproteínas VLDL/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Células Cultivadas , Proteínas de la Membrana/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proproteína Convertasa 9/deficiencia , Proproteína Convertasa 9/metabolismo , Receptores de LDL/deficiencia , Receptores de LDL/metabolismo
10.
Neurobiol Dis ; 151: 105257, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33434616

RESUMEN

TGFß-activated kinase 1 (TAK1) is a master regulator that drives multiple cell death and proinflammatory signaling pathways, making it a promising therapeutic target to treat ischemic stroke. However, whether targeting TAK1 could improve stroke outcomes has never been tested in female subjects, hindering its potential translation into clinical use. Here we examined the therapeutic effect of 5Z-7-Oxozeaenol (OZ), a selective TAK1 inhibitor, in ovariectomized female mice after middle cerebral artery occlusion (MCAO). OZ significantly reduced neuronal cell death and axonal injury at the acute stage and mitigated neuroinflammation at the subacute stage after MCAO in ovariectomized female mice. Consistent with RNA sequencing analysis that TAK1 activation contributed to microglia/macrophage-mediated inflammatory responses in the post-stroke brain, inhibition of TAK1 with OZ caused phenotypic shift of microglia/macrophages toward an inflammation-resolving state. Furthermore, microglia/macrophage-specific TAK1 knockout (TAK1 mKO) reproduced OZ's effects, causally confirming the role of TAK1 in determining proinflammatory microglial/macrophage responses in post-stroke females. Post-stroke treatment with OZ for 5 days effectively promoted long-term neurological recovery and the integrity of both gray matter and white matter in female mice. Together, the TAK1 inhibitor OZ elicits long-lasting improvement of stroke outcomes in female mice, at least partially through enhancing beneficial microglial/macrophage responses and inflammation resolution. Given its therapeutic efficacy on both male and female rodents, TAK1 inhibitor is worth further investigation as a valid treatment to ischemic stroke.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Infarto de la Arteria Cerebral Media/metabolismo , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Macrófagos/metabolismo , Microglía/metabolismo , Recuperación de la Función/efectos de los fármacos , Animales , Femenino , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovariectomía , Zearalenona/análogos & derivados , Zearalenona/farmacología
11.
Nat Chem Biol ; 14(7): 715-722, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29915236

RESUMEN

ATP-binding cassette (ABC) transporters use the energy of ATP hydrolysis to move molecules through cellular membranes. They are directly linked to human diseases, cancer multidrug resistance, and bacterial virulence. Very little is known of the conformational dynamics of ABC transporters, especially at the single-molecule level. Here, we combine single-molecule spectroscopy and a novel molecular simulation approach to investigate the conformational dynamics of the ABC transporter BtuCD. We observe a single dominant population of molecules in each step of the transport cycle and tight coupling between conformational transitions and ligand binding. We uncover transient conformational changes that allow substrate to enter the transporter. This is followed by a 'squeezing' motion propagating from the extracellular to the intracellular side of the translocation cavity. This coordinated sequence of events provides a mechanism for the unidirectional transport of vitamin B12 by BtuCD.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/química , Cisteína/química , Proteínas de Escherichia coli/química , Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas de Escherichia coli/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Modelos Moleculares , Conformación Proteica
12.
Proc Natl Acad Sci U S A ; 114(38): 10089-10094, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28878024

RESUMEN

Sugar Will Eventually be Exported Transporters (SWEETs) are recently identified sugar transporters that can discriminate and transport di- or monosaccharides across a membrane following the concentration gradient. SWEETs play key roles in plant biological processes, such as pollen nutrition, nectar secretion, seed filling, and phloem loading. SWEET13 from Arabidopsis thaliana (AtSWEET13) is an important sucrose transporter in pollen development. Here, we report the 2.8-Å resolution crystal structure of AtSWEET13 in the inward-facing conformation with a substrate analog, 2'-deoxycytidine 5'-monophosphate, bound in the central cavity. In addition, based on the results of an in-cell transport activity assay and single-molecule Förster resonance energy transfer analysis, we suggest a mechanism for substrate selectivity based on the size of the substrate-binding pocket. Furthermore, AtSWEET13 appears to form a higher order structure presumably related to its function.


Asunto(s)
Proteínas de Arabidopsis/química , Arabidopsis/química , Proteínas de Transporte de Membrana/química , Arabidopsis/genética , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Desoxicitidina Monofosfato , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Relación Estructura-Actividad
13.
Biophys J ; 116(12): 2296-2303, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31146923

RESUMEN

Bacterial multidrug-resistance transporters of the major facilitator superfamily are distinguished by their extraordinary ability to bind structurally diverse substrates, thus serving as a highly efficient tool to protect cells from multiple toxic substances present in their environment, including antibiotic drugs. However, details of the dynamic conformational changes of the transport cycle involved remain to be elucidated. Here, we used the single-molecule fluorescence resonance energy transfer technique to investigate the conformational behavior of the Escherichia coli multidrug transporter MdfA under conditions of different substrates, pH, and alkali metal ions. Our data show that different substrates exhibit distinct effects on both the conformational distribution and transition rate between two major conformations. Although the cationic substrate tetraphenylphosphonium favors the outward-facing conformation, it has less effect on the transition rate. In contrast, binding of the electroneutral substrate chloramphenicol tends to stabilize the inward-facing conformation and decreases the transition rate. Therefore, our study supports the notion that the MdfA transporter uses distinct mechanisms to transport electroneutral and cationic substrates.


Asunto(s)
Resistencia a Múltiples Medicamentos , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli , Transferencia Resonante de Energía de Fluorescencia , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/metabolismo , Concentración de Iones de Hidrógeno , Modelos Moleculares , Potasio/farmacología , Conformación Proteica
14.
Biochemistry ; 58(14): 1931-1941, 2019 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-30888187

RESUMEN

The chaperone-usher secretion pathway is a conserved bacterial protein secretion system dedicated to the biogenesis of adhesive fibers. Usher, a multidomain-containing outer membrane protein, plays a central role in this process by acting as a molecular machine that recruits different chaperone-subunit complexes, catalyzes subunit polymerization, and forms a channel for secretion of the assembled subunits. While recent crystal structural studies have greatly advanced our understanding of the structure and function of ushers, the overall architecture of the full-length apo-usher, the molecular events that dictate conformational changes in usher during pilus biogenesis, and its activation by the specific chaperone-adhesin complex remain largely elusive. Using single-molecule fluorescence resonance energy transfer studies, we found that the substrate-free usher FimD (apo-FimD) adopts a contracted conformation that is distinct from its substrate-bound states; both the N-terminal domain (NTD) and the C-terminal domain (CTD) of apo-FimD are highly dynamic, and FimD coordinates its domain conformational changes via intramolecular domain conformation signaling. By combining these studies with in vitro photo-cross-linking studies, we further show that only the chaperone-bound adhesin (FimC:FimH) can be transferred to the CTD, dislocates the plug domain, and triggers conformational changes in the remaining FimD domains. Taken together, these studies delineate an overall architecture of the full-length apo-FimD, provide detailed mechanic insight into the activation of apo-FimD, and explain why FimD could adjust its conformational states to perform multiple functions in each cycle of pilus subunit addition and ensure that pilus assembly proceeds progressively in a cellular energy-free environment.


Asunto(s)
Proteínas de Escherichia coli/química , Proteínas Fimbrias/química , Transferencia Resonante de Energía de Fluorescencia/métodos , Chaperonas Moleculares/química , Conformación Proteica , Dominios Proteicos , Sistemas de Secreción Bacterianos/genética , Sistemas de Secreción Bacterianos/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas Fimbrias/genética , Proteínas Fimbrias/metabolismo , Polarización de Fluorescencia , Cinética , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Mutación , Unión Proteica , Transducción de Señal/genética
15.
Proc Natl Acad Sci U S A ; 113(46): 13015-13020, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27803324

RESUMEN

STAT6 participates in classical IL-4/IL-13 signaling and stimulator of interferon genes-mediated antiviral innate immune responses. Aberrations in STAT6-mediated signaling are linked to development of asthma and diseases of the immune system. In addition, STAT6 remains constitutively active in multiple types of cancer. Therefore, targeting STAT6 is an attractive proposition for treating related diseases. Although a lot is known about the role of STAT6 in transcriptional regulation, molecular details on how STAT6 recognizes and binds specific segments of DNA to exert its function are not clearly understood. Here, we report the crystal structures of a homodimer of phosphorylated STAT6 core fragment (STAT6CF) alone and bound with the N3 and N4 DNA binding site. Analysis of the structures reveals that STAT6 undergoes a dramatic conformational change on DNA binding, which was further validated by performing molecular dynamics simulation studies and small angle X-ray scattering analysis. Our data show that a larger angle at the intersection where the two protomers of STAT meet and the presence of a unique residue, H415, in the DNA-binding domain play important roles in discrimination of the N4 site DNA from the N3 site by STAT6. H415N mutation of STAT6CF decreased affinity of the protein for the N4 site DNA, but increased its affinity for N3 site DNA, both in vitro and in vivo. Results of our structure-function studies on STAT6 shed light on mechanism of DNA recognition by STATs in general and explain the reasons underlying STAT6's preference for N4 site DNA over N3.


Asunto(s)
ADN/metabolismo , Factor de Transcripción STAT6/química , Factor de Transcripción STAT6/metabolismo , Sitios de Unión , Cristalización , ADN/química , Escherichia coli/genética , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Mutación , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Factor de Transcripción STAT6/genética
16.
Biochem Biophys Res Commun ; 491(3): 603-608, 2017 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-28760338

RESUMEN

The heterotrimeric G proteins (Gαßγ) act as molecular switches to mediate signal transduction from G protein-coupled receptors to downstream effectors. Upon interaction with an activated receptor, G protein exchanges its bound GDP with GTP, stimulating downstream signal transmission. Release of GDP requires a structural rearrangement between the GTPase domain and helical domain of the Gα subunit. Here, we used single molecule fluorescence resonance energy transfer (smFRET) technique to study the conformational dynamics of these two domains in the apo state and in the binding of different ligands. Direct imaging of individual molecules showed that the Giα subunit is highly dynamic, and at least three major conformations of Giα could be observed in the apo state. Upon binding of GDP, Giα becomes dramatically less dynamic, resulting in a closed conformation between the two domains. We postulate that changes between the three conformations are sequential, and the three conformations appear to have distinct affinities toward GDP.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia/métodos , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/química , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/ultraestructura , Guanosina Difosfato/química , Imagen Molecular/métodos , Sitios de Unión , Activación Enzimática , Unión Proteica , Conformación Proteica , Dominios Proteicos , Subunidades de Proteína
17.
Nature ; 474(7349): 109-13, 2011 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-21516104

RESUMEN

Neurotransmitter/Na(+) symporters (NSSs) terminate neuronal signalling by recapturing neurotransmitter released into the synapse in a co-transport (symport) mechanism driven by the Na(+) electrochemical gradient. NSSs for dopamine, noradrenaline and serotonin are targeted by the psychostimulants cocaine and amphetamine, as well as by antidepressants. The crystal structure of LeuT, a prokaryotic NSS homologue, revealed an occluded conformation in which a leucine (Leu) and two Na(+) are bound deep within the protein. This structure has been the basis for extensive structural and computational exploration of the functional mechanisms of proteins with a LeuT-like fold. Subsequently, an 'outward-open' conformation was determined in the presence of the inhibitor tryptophan, and the Na(+)-dependent formation of a dynamic outward-facing intermediate was identified using electron paramagnetic resonance spectroscopy. In addition, single-molecule fluorescence resonance energy transfer imaging has been used to reveal reversible transitions to an inward-open LeuT conformation, which involve the movement of transmembrane helix TM1a away from the transmembrane helical bundle. We investigated how substrate binding is coupled to structural transitions in LeuT during Na(+)-coupled transport. Here we report a process whereby substrate binding from the extracellular side of LeuT facilitates intracellular gate opening and substrate release at the intracellular face of the protein. In the presence of alanine, a substrate that is transported ∼10-fold faster than leucine, we observed alanine-induced dynamics in the intracellular gate region of LeuT that directly correlate with transport efficiency. Collectively, our data reveal functionally relevant and previously hidden aspects of the NSS transport mechanism that emphasize the functional importance of a second substrate (S2) binding site within the extracellular vestibule. Substrate binding in this S2 site appears to act cooperatively with the primary substrate (S1) binding site to control intracellular gating more than 30 Šaway, in a manner that allows the Na(+) gradient to power the transport mechanism.


Asunto(s)
Activación del Canal Iónico/efectos de los fármacos , Modelos Moleculares , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/metabolismo , Humanos , Leucina/metabolismo , Litio/metabolismo , Mutación , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/química , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/genética , Unión Proteica/genética , Estructura Secundaria de Proteína , Sodio/metabolismo , Sodio/farmacología
18.
Cell Physiol Biochem ; 38(6): 2375-88, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27300751

RESUMEN

BACKGROUND/AIMS: Bone marrow-derived mesenchymal stem cells (MSCs) are responsible for new bone formation during adulthood. Accumulating evidences showed that Osthole promotes the osteogenic differentiation in primary osteoblasts. The aim of this study was to investigate whether Osthole exhibits a potential to stimulate the osteogenic differentiation of MSCs and the underlying mechanism. METHODS: MSCs were treated with a gradient concentration of Osthole (6.25 µM, 12.5 µM, and 25 µM). Cell proliferation was assessed by western blotting with the proliferating cell nuclear antigen (PCNA) and Cyclin D1 antibodies, fluorescence activated cell sorting (FACS), and cell counting kit 8 (CCK8). MSCs were cultured in osteogenesis-induced medium for one or two weeks. The osteogenic differentiation of MSCs was estimated by Alkaline Phosphatase (ALP) staining, Alizarin red staining, Calcium influx, and quantitative PCR (qPCR). The underlying mechanism of Osthole-induced osteogenesis was further evaluated by western blotting with antibodies in Wnt/ß-catenin, PI3K/Akt, BMPs/smad1/5/8, and MAPK signaling pathways. RESULTS: Osthole inhibited proliferation of rat MSCs in a dose-dependent manner. Osthole suppressed osteogenic differentiation of rat MSCs by down-regulating the activities of Wnt/ß-catenin and Erk1/2-MAPK signaling. CONCLUSIONS: Osthole inhibits the proliferation and osteogenic differentiation of rat MSCs, which might be mediated through blocking the Wnt/ß-catenin and Erk1/2-MAPK signaling pathways.


Asunto(s)
Cumarinas/farmacología , Medicamentos Herbarios Chinos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Fosfatasa Alcalina/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Ratas , beta Catenina/metabolismo
19.
Nature ; 465(7295): 188-93, 2010 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-20463731

RESUMEN

Neurotransmitter:Na(+) symporters (NSS) remove neurotransmitters from the synapse in a reuptake process that is driven by the Na(+) gradient. Drugs that interfere with this reuptake mechanism, such as cocaine and antidepressants, profoundly influence behaviour and mood. To probe the nature of the conformational changes that are associated with substrate binding and transport, we have developed a single-molecule fluorescence imaging assay and combined it with functional and computational studies of the prokaryotic NSS homologue LeuT. Here we show molecular details of the modulation of intracellular gating of LeuT by substrates and inhibitors, as well as by mutations that alter binding, transport or both. Our direct observations of single-molecule transitions, reflecting structural dynamics of the intracellular region of the transporter that might be masked by ensemble averaging or suppressed under crystallographic conditions, are interpreted in the context of an allosteric mechanism that couples ion and substrate binding to transport.


Asunto(s)
Aquifoliaceae/química , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/química , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/metabolismo , Alanina/metabolismo , Regulación Alostérica , Proteínas Bacterianas/genética , Cristalografía por Rayos X , Cisteína/química , Cisteína/metabolismo , Escherichia coli , Transferencia Resonante de Energía de Fluorescencia , Leucina/metabolismo , Modelos Moleculares , Simulación de Dinámica Molecular , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/genética , Conformación Proteica , Sodio/metabolismo
20.
Mol Cell ; 30(6): 667-77, 2008 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-18570870

RESUMEN

Eukaryotic neurotransmitter:sodium symporters (NSSs), targets for antidepressants and psychostimulants, terminate neurotransmission by sodium-driven reuptake. The crystal structure of LeuT(Aa), a prokaryotic NSS homolog, revealed an occluded state in which one leucine and two Na(+) ions are bound, but provided limited clues to the molecular mechanism of transport. Using steered molecular dynamics simulations, we explored the substrate translocation pathway of LeuT. We identified a second substrate binding site located in the extracellular vestibule comprised of residues shown recently to participate in binding tricyclic antidepressants. Binding and flux experiments showed that the two binding sites can be occupied simultaneously. The substrate in the secondary site allosterically triggers intracellular release of Na(+) and substrate from the primary site, thereby functioning as a "symport effector." Because tricyclic antidepressants bind differently to this secondary site, they do not promote substrate release from the primary site and thus act as symport uncouplers and inhibit transport.


Asunto(s)
Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/metabolismo , Sodio/metabolismo , Sitios de Unión , Plaquetas/metabolismo , Comunicación Celular , Simulación por Computador , Humanos , Cinética , Leucina/química , Leucina/metabolismo , Modelos Moleculares , Mutagénesis , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/química , Conformación Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Cloruro de Sodio/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA