Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(35): e2400194121, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39172792

RESUMEN

Size-dependent phagocytosis is a well-characterized phenomenon in monocytes and macrophages. However, this size effect for preferential gene delivery to these important cell targets has not been fully exploited because commonly adopted stabilization methods for electrostatically complexed nucleic acid nanoparticles, such as PEGylation and charge repulsion, typically arrest the vehicle size below 200 nm. Here, we bridge the technical gap in scalable synthesis of larger submicron gene delivery vehicles by electrostatic self-assembly of charged nanoparticles, facilitated by a polymer structurally designed to modulate internanoparticle Coulombic and van der Waals forces. Specifically, our strategy permits controlled assembly of small poly(ß-amino ester)/messenger ribonucleic acid (mRNA) nanoparticles into particles with a size that is kinetically tunable between 200 and 1,000 nm with high colloidal stability in physiological media. We found that assembled particles with an average size of 400 nm safely and most efficiently transfect monocytes following intravenous administration and mediate their differentiation into macrophages in the periphery. When a CpG adjuvant is co-loaded into the particles with an antigen mRNA, the monocytes differentiate into inflammatory dendritic cells and prime adaptive anticancer immunity in the tumor-draining lymph node. This platform technology offers a unique ligand-independent, particle-size-mediated strategy for preferential mRNA delivery and enables therapeutic paradigms via monocyte programming.


Asunto(s)
Monocitos , Nanopartículas , ARN Mensajero , Monocitos/metabolismo , Nanopartículas/química , ARN Mensajero/genética , ARN Mensajero/metabolismo , Animales , Ratones , Humanos , Polielectrolitos/química , Macrófagos/metabolismo , Poliaminas/química , Tamaño de la Partícula , Diferenciación Celular , Técnicas de Transferencia de Gen , Células Dendríticas/metabolismo , Electricidad Estática , Polímeros
2.
Biochem Biophys Res Commun ; 630: 133-142, 2022 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-36155059

RESUMEN

The phenotypic transformation of vascular smooth muscle cells (VSMCs) plays a key role in the pathological process of atherosclerosis (AS), and TRPM7 is involved in this process. In this study, we verified whether circRNAs participate in the phenotypic transformation of VSMCs by regulating TRPM7 in AS. The RNA-sequencing data of atherosclerosis were downloaded and analysed from the GEO database. Only hsa_circ_0021155 related to TRPM7 was differentially expressed in AS. circRNA distribution and expression were observed via FISH and PCR. CCK8, scratch test and Transwell assay were used to observe the proliferation and migration of cells. Western blot was performed to examine changes in α-actin, calponin, SMMHC and TRPM7 proteins. The expression of hsa_circ_0021155 against has-miR-4459/miR-3689c was verified via PCR. The ceRNA relationship of TPRM7-miR4459-circ0021155 was verified via dual luciferase assay, and the effects of miR4459 mimic/inhibitor on the proliferation of cells were further observed. The expression of hsa_circ_0021155 and OX-LDL was increased in VSMCs. hsa_circ_0021155 promoted the expression of TRPM7 and inhibited the protein expression of α-actin, calponin and SMMHC. In addition, it promoted the proliferation and migration of cells and inhibited the expression of miR-3689c and miR-4459 but did not affect miR-4756-5p. The dual luciferase assay showed that circ0021155-miR4459-TRPM7 mRNA was highly compatible and could be mutually regulated by a ceRNA network. In conclusion, hsa_circ_0021155 regulates the proliferation, migration and phenotype transformation of VSMCs induced by OX-LDL via the miR-4459/TRPM7 axis. hsa_circ_0021155 and TRPM7 may offer novel therapeutic targets for atherosclerosis.


Asunto(s)
Aterosclerosis , MicroARNs , Canales Catiónicos TRPM , Actinas/metabolismo , Apoptosis/genética , Aterosclerosis/genética , Aterosclerosis/patología , Movimiento Celular/genética , Proliferación Celular/genética , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Músculo Liso Vascular/metabolismo , Fenotipo , Proteínas Serina-Treonina Quinasas , ARN Circular/genética , ARN Mensajero , Canales Catiónicos TRPM/genética
3.
Cell Biochem Funct ; 39(7): 908-920, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34296452

RESUMEN

Increasing attention has been paid on the application of biodegradable materials such as magnesium and its alloys in neuron repair. AZ91D magnesium alloy coated with carbon nanotubes (CNTs) and/or calcium phosphate (CaP)/chitosan (CS) was fabricated in this study. To evaluate the bioactivity of these AZ91D-based composites, the extracts were prepared by immersing samples in modified simulated body fluid (m-SBF) for 0, 2, 8, 16, 24, 34, 44, 60, or 90 days. Immunofluorescence staining for neuronal class III ß-tubulin (TUJ1) revealed that both CNTs-CaP/CS-AZ91D and CaP/CS-AZ91D extracts promoted axon outgrowth of dorsal root ganglia (DRG) neurons, accompanied with increased expression of phosphorylated focal adhesion kinase (p-FAK) and growth associated protein-43 (GAP-43). Besides, the extracts increased the expression and the release of neurotrophic factors including nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF). ERK signalling was activated in DRG neurons after treating with either CNTs-CaP/CS-AZ91D or CaP/CS-AZ91D extracts, and its inhibition with U0126 counteracted the beneficial effects of these extracts on DRG neuron. Overall, the extracts from these AZ91D-based composites might promote DRG neuron growth via activating ERK signalling pathway. Notably, CNTs-CaP/CS-AZ91D extracts showed a better promoting effect on neuron growth than CaP/CS-AZ91D. Assessment of ion elements showed that the addition of CNTs coating enhanced magnesium corrosion resistance and reduced the deposition of calcium and phosphorus on the surface of CaP/CS-AZ91D alloy. These findings demonstrate that CNTs-CaP/CS-AZ91D likely provide a more suitable environment for neuron growth, which suggests a potential implantable biomaterial for the treatment of nerve injury. SIGNIFICANCE: AZ91D magnesium alloy coated with carbon nanotubes (CNTs) and/or calcium phosphate (CaP)/chitosan (CS) was fabricated and their immersion extracts were prepared using modified simulated body fluid in this study. Both extracts from CNTs-CaP/CS and CaP/CS-coated AZ91D magnesium alloy promotes rat dorsal root ganglia (DRG) neuron growth via activating ERK signalling pathway. Notably, the addition of CNTs improves the performance of CaP/CS-AZ91D. For the first time, our research demonstrates that CNTs-CaP/CS-AZ91D likely provide a suitable environment for neuron growth, suggesting these AZ91D-based composites as potential implantable biomaterials for the treatment of nerve injury.


Asunto(s)
Aleaciones/farmacología , Fosfatos de Calcio/farmacología , Quitosano/farmacología , Magnesio/farmacología , Nanotubos de Carbono/química , Aleaciones/química , Aleaciones/aislamiento & purificación , Animales , Fosfatos de Calcio/química , Fosfatos de Calcio/aislamiento & purificación , Quitosano/química , Quitosano/aislamiento & purificación , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/crecimiento & desarrollo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Magnesio/química , Magnesio/aislamiento & purificación , Neuronas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
4.
Hum Mol Genet ; 27(4): 625-637, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29294000

RESUMEN

Paroxysmal kinesigenic dyskinesia (PKD) is a heterogeneous movement disorder characterized by recurrent dyskinesia attacks triggered by sudden movement. PRRT2 has been identified as the first causative gene of PKD. However, it is only responsible for approximately half of affected individuals, indicating that other loci are most likely involved in the etiology of this disorder. To explore the underlying causative gene of PRRT2-negative PKD, we used a combination strategy including linkage analysis, whole-exome sequencing and copy number variations analysis to detect the genetic variants within a family with PKD. We identified a linkage locus on chromosome 12 (12p13.32-12p12.3) and detected a novel heterozygous mutation c.956 T>G (p.319 L>R) in the potassium voltage-gated channel subfamily A member 1, KCNA1. Whole-exome sequencing in another 58 Chinese patients with PKD who lacked mutations in PRRT2 revealed another novel mutation in the KCNA1 gene [c.765 C>A (p.255 N>K)] within another family. Biochemical analysis revealed that the L319R mutant accelerated protein degradation via the proteasome pathway and disrupted membrane expression of the Kv1.1 channel. Electrophysiological examinations in transfected HEK293 cells showed that both the L319R and N255K mutants resulted in reduced potassium currents and respective altered gating properties, with a dominant negative effect on the Kv1.1 wild-type channel. Our study suggests that these mutations in KCNA1 cause the Kv1.1 channel dysfunction, which leads to familial PKD. The current study further extended the genotypic spectrum of this disorder, indicating that Kv1.1 channel dysfunction maybe one of the underlying defects in PKD.


Asunto(s)
Distonía/genética , Canal de Potasio Kv.1.1/genética , Adulto , Pueblo Asiatico , Variaciones en el Número de Copia de ADN , Femenino , Células HEK293 , Humanos , Masculino , Persona de Mediana Edad , Mutación/genética , Linaje
5.
Cell Physiol Biochem ; 40(6): 1274-1288, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27997885

RESUMEN

BACKGROUND: Streptozotocin (STZ) has served as an agent to generate an Alzheimer's disease (AD) model in rats, while edaravone (EDA), a novel free radical scavenger, has recently emerged as an effective treatment for use in vivo and vitro AD models. However, to date, these beneficial effects of EDA have only been clearly demonstrated within STZ-induced animal models of AD and in cell models of AD. A better understanding of the mechanisms of EDA may provide the opportunity for their clinical application in the treatment of AD. Therefore, the purpose of this study was to investigate the underlying mechanisms of STZ and EDA as assessed upon electrophysiological alterations in CA1 pyramidal neurons of rat hippocampal slices. METHODS: Through measures of evoked excitatory postsynaptic currents (eEPSCs), AMPAR-mediated eEPSCs (eEPSCsAMPA), evoked inhibitory postsynaptic currents (eIPSCs), evoked excitatory postsynaptic current paired pulse ratio (eEPSC PPR) and evoked inhibitory postsynaptic current paired pulse ratio (eIPSC PPR), it was possible to investigate mechanisms as related to the neurotoxicity of STZ and reductions in these effects by EDA. RESULTS: Our results showed that STZ (1000 µM) significantly inhibited peak amplitudes of eEPSCs, eEPSCsAMPA and eIPSCs, while EDA (1000 µM) attenuated these STZ-induced changes at holding potentials ranging from -60mV to +40 mV for EPSCs and -60mV to +20 mV for IPSCs. Our work also indicated that mean eEPSC PPR were substantially altered by STZ, effects which were partially restored by EDA. In contrast, no significant effects upon eIPSC PPR were obtained in response to STZ and EDA. CONCLUSION: Our data suggest that STZ inhibits glutamatergic transmission involving pre-synaptic mechanisms and AMPAR, and that STZ inhibits GABAergic transmission by post-synaptic mechanisms within CA1 pyramidal neurons. These effects are attenuated by EDA.


Asunto(s)
Antipirina/análogos & derivados , Región CA1 Hipocampal/citología , Células Piramidales/fisiología , Estreptozocina/farmacología , Transmisión Sináptica/efectos de los fármacos , Animales , Antipirina/farmacología , Edaravona , Capacidad Eléctrica , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Femenino , Masculino , Potenciales de la Membrana/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Ratas Wistar , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/farmacología
7.
Biochem Biophys Res Commun ; 463(4): 1297-304, 2015 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-26093297

RESUMEN

PURPOSE: Genistein, as a protein tyrosine kinase inhibitor, has been shown to possess anti-atherosclerotic effects. Since the smooth muscle cell-derived foam cells are key components of atherosclerotic plaques. The aim of this study is to investigate the influence of genistein on foam cell transformation from vascular smooth muscle cells and possible mechanisms contributing to these effects. METHODS AND RESULTS: Vascular smooth muscle cells exposed to ox-LDL developed into foam cell, as demonstrated by Oil Red O staining and cholesterol content analysis. Ox-LDL induced phenotype transformation of smooth muscle cells, decreased expression of α-actin and increased expression of CD68 (a specific marker for monocytes, can also function as a subtype of scavenger receptors). The expression of scavenger receptors CD36 and LOX-1 was measured, and their role in foam cell formation in the presence of genistein, daidzein (a structurally similar analogue of genistein) and herbimycin A (a commonly tyrosine kinase inhibitor). The results showed that foam cell formation was markedly reduced by genistein and herbimycin A, as well as the expression of CD68, CD36 and LOX-1. However, daidzein had no such effect. In addition, genistein-induced down-regulation of CD68, CD36 and LOX-1 could be reversed by sodium orthovanadate (a membrane-permeable protein tyrosine phosphatase inhibitor). CONCLUSION: The results showed that ox-LDL induce smooth muscle cell-derived foam cell formation and transform the phenotype of smooth muscle cell. While tyrosine kinase inhibitor, genistein could suppress smooth muscle cell-derived foam cell formation through inhibiting the protein expressions of CD68, CD36 and LOX-1.


Asunto(s)
Genisteína/farmacología , Músculo Liso Vascular/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/metabolismo , Animales , Células Cultivadas , Isoflavonas/farmacología , Lipoproteínas LDL/farmacología , Masculino , Músculo Liso Vascular/citología , Músculo Liso Vascular/enzimología , Ratas , Ratas Wistar , Rifabutina/análogos & derivados , Rifabutina/farmacología
8.
Mol Cell Biochem ; 406(1-2): 139-49, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25987498

RESUMEN

Arterial calcification is common in vascular diseases and involves conversion of vascular smooth muscle cells (VSMCs) to an osteoblast phenotype. Clinical studies suggest that the development of atherosclerosis can be promoted by homocysteine (HCY), but the mechanisms remain unclear. Here, we determined whether increases in HCY levels lead to an increase in VSMC calcification and differentiation, and examined the role of an extracellular matrix remodeler, matrix metalloproteinase-2 (MMP-2). Rat VSMCs were exposed to calcification medium in the absence or presence of HCY (10, 100 or 200 µmol/L) or an MMP-2 inhibitor (10(-6) or 10(-5) mol/L). MTT assays were performed to determine the cytotoxicity of the MMP-2 inhibitor in calcification medium containing 200 µmol/L HCY. Calcification was assessed by measurements of calcium deposition and alkaline phosphatase (ALP) activity as well as von Kossa staining. Expression of osteocalcin, bone morphogenetic protein (BMP)-2, and osteopontin, and MMP-2 was determined by immunoblotting. Calcification medium induced osteogenic differentiation of VSMCs. HCY promoted calcification, increased osteocalcin and BMP-2 expression, and decreased expression of osteopontin. MMP-2 expression was increased by HCY in a dose-dependent manner in VSMCs exposed to both control and calcification medium. The MMP-2 inhibitor decreased the calcium content and ALP activity, and attenuated the osteoblastic phenotype of VSMCs. Vascular calcification and osteogenic differentiation of VSMCs were positively regulated by HCY through increased/restored MMP-2 expression, increased expression of calcification proteins, and decreased anti-calcification protein levels. In summary, MMP-2 inhibition may be a protective strategy against VSMC calcification.


Asunto(s)
Diferenciación Celular , Homocisteína/farmacología , Metaloproteinasa 2 de la Matriz/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/patología , Calcificación Vascular/enzimología , Animales , Fosfatos de Calcio/metabolismo , Células Cultivadas , Inducción Enzimática , Masculino , Metaloproteinasa 2 de la Matriz/genética , Músculo Liso Vascular/metabolismo , Osteogénesis , Fenotipo , Ratas , Ratas Wistar
9.
J Cardiovasc Pharmacol ; 66(1): 16-24, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26164719

RESUMEN

Transient receptor potential melastatin 7 (TRPM7) is a Ca, Mg permeable nonselective cation channel of the TRP channel superfamily and plays an important role in cell growth and proliferation. Compounds that alter the activity and expression of the channel protein might be of therapeutic interest. In this study, we investigated the effects of genistein on TRPM7 channels and the proliferation of rat aortic smooth muscle cells (RAoSMCs). In primary cultured RAoSMCs, acute genistein (50 µM) exposure inhibited native TRPM7 currents, whereas chronic expose to genistein (50 µM) downregulated TRPM7 protein expression. The downregulation of TRPM7 protein expression induced by genistein was mimicked by c-Src inhibitor (PP2), but not by epidermal growth factor receptor tyrosine kinase inhibitor (lavendustin A), or daidzein. Additionally, genistein (50 µM) attenuated angiotensin II-induced cell proliferation. This study is the first to demonstrate inhibition of TRPM7 by isoflavone genistein through c-Src tyrosine kinase inhibition in RAoSMCs. Our results not only provide a new modulation mechanism of TRPM7 but also suggest that TRPM7 may serve as a new therapeutic target of genistein in the treatment of vascular diseases.


Asunto(s)
Angiotensina II/toxicidad , Aorta/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Genisteína/farmacología , Miocitos del Músculo Liso/efectos de los fármacos , Canales Catiónicos TRPM/antagonistas & inhibidores , Animales , Aorta/metabolismo , Proliferación Celular/fisiología , Células Cultivadas , Masculino , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Ratas , Ratas Wistar , Canales Catiónicos TRPM/metabolismo
10.
Mol Ther Methods Clin Dev ; 32(1): 101194, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38352269

RESUMEN

The transfection efficiency and stability of the delivery vehicles of plasmid DNA (pDNA) are critical metrics to ensure high-quality and high-yield production of viral vectors. We previously identified that the optimal size of pDNA/poly(ethylenimine) (PEI) transfection particles is 400-500 nm and developed a bottom-up assembly method to construct stable 400-nm pDNA/PEI particles and benchmarked their transfection efficiency in producing lentiviral vectors (LVVs). Here, we report scale-up production protocols for such transfection particles. Using a two-inlet confined impinging jet (CIJ) mixer with a dual syringe pump set-up, we produced a 1-L batch at a flow rate of 100 mL/min, and further scaled up this process with a larger CIJ mixer and a dual peristaltic pump array, allowing for continuous production at a flow rate of 1 L/min without a lot size limit. We demonstrated the scalability of this process with a 5-L lot and validated the quality of these 400-nm transfection particles against the target product profile, including physical properties, shelf and on-bench stability, transfection efficiency, and LVV production yield in both 15-mL bench culture and 2-L bioreactor runs. These results confirm the potential of this particle assembly process as a scalable manufacturing platform for viral vector production.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA