Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 179
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Traffic ; 23(10): 496-505, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36054788

RESUMEN

Localization of mRNA facilitates spatiotemporally controlled protein expression in neurons. In axons, mRNA transport followed by local protein synthesis plays a critical role in axonal growth and guidance. However, it is not yet clearly understood how mRNA is transported to axonal subcellular sites and what regulates axonal mRNA localization. Using a transgenic mouse model in which endogenous ß-actin mRNA is fluorescently labeled, we investigated ß-actin mRNA movement in axons of hippocampal neurons. We cultured neurons in microfluidic devices to separate axons from dendrites and performed single-particle tracking of axonal ß-actin mRNA. Compared with dendritic ß-actin mRNA, axonal ß-actin mRNA showed less directed motion and exhibited mostly subdiffusive motion, especially near filopodia and boutons in mature dissociated hippocampal neurons. We found that axonal ß-actin mRNA was likely to colocalize with actin patches (APs), regions that have a high density of filamentous actin (F-actin) and are known to have a role in branch initiation. Moreover, simultaneous imaging of F-actin and axonal ß-actin mRNA in live neurons revealed that moving ß-actin mRNA tended to be docked in the APs. Our findings reveal that axonal ß-actin mRNA localization is facilitated by actin networks and suggest that localized ß-actin mRNA plays a potential role in axon branch formation.


Asunto(s)
Actinas , Axones , Actinas/metabolismo , Animales , Axones/metabolismo , Células Cultivadas , Hipocampo/metabolismo , Ratones , Neuronas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
2.
Circ Res ; 131(2): e2-e21, 2022 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-35701867

RESUMEN

BACKGROUND: Mutations in PIEZO1 (Piezo type mechanosensitive ion channel component 1) cause human lymphatic malformations. We have previously uncovered an ORAI1 (ORAI calcium release-activated calcium modulator 1)-mediated mechanotransduction pathway that triggers lymphatic sprouting through Notch downregulation in response to fluid flow. However, the identity of its upstream mechanosensor remains unknown. This study aimed to identify and characterize the molecular sensor that translates the flow-mediated external signal to the Orai1-regulated lymphatic expansion. METHODS: Various mutant mouse models, cellular, biochemical, and molecular biology tools, and a mouse tail lymphedema model were employed to elucidate the role of Piezo1 in flow-induced lymphatic growth and regeneration. RESULTS: Piezo1 was found to be abundantly expressed in lymphatic endothelial cells. Piezo1 knockdown in cultured lymphatic endothelial cells inhibited the laminar flow-induced calcium influx and abrogated the flow-mediated regulation of the Orai1 downstream genes, such as KLF2 (Krüppel-like factor 2), DTX1 (Deltex E3 ubiquitin ligase 1), DTX3L (Deltex E3 ubiquitin ligase 3L,) and NOTCH1 (Notch receptor 1), which are involved in lymphatic sprouting. Conversely, stimulation of Piezo1 activated the Orai1-regulated mechanotransduction in the absence of fluid flow. Piezo1-mediated mechanotransduction was significantly blocked by Orai1 inhibition, establishing the epistatic relationship between Piezo1 and Orai1. Lymphatic-specific conditional Piezo1 knockout largely phenocopied sprouting defects shown in Orai1- or Klf2- knockout lymphatics during embryo development. Postnatal deletion of Piezo1 induced lymphatic regression in adults. Ectopic Dtx3L expression rescued the lymphatic defects caused by Piezo1 knockout, affirming that the Piezo1 promotes lymphatic sprouting through Notch downregulation. Consistently, transgenic Piezo1 expression or pharmacological Piezo1 activation enhanced lymphatic sprouting. Finally, we assessed a potential therapeutic value of Piezo1 activation in lymphatic regeneration and found that a Piezo1 agonist, Yoda1, effectively suppressed postsurgical lymphedema development. CONCLUSIONS: Piezo1 is an upstream mechanosensor for the lymphatic mechanotransduction pathway and regulates lymphatic growth in response to external physical stimuli. Piezo1 activation presents a novel therapeutic opportunity for preventing postsurgical lymphedema. The Piezo1-regulated lymphangiogenesis mechanism offers a molecular basis for Piezo1-associated lymphatic malformation in humans.


Asunto(s)
Vasos Linfáticos , Linfedema , Animales , Células Endoteliales/metabolismo , Humanos , Canales Iónicos/genética , Canales Iónicos/metabolismo , Vasos Linfáticos/metabolismo , Linfedema/metabolismo , Mecanotransducción Celular/fisiología , Ratones , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
3.
Nat Mater ; 20(1): 100-107, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32807919

RESUMEN

The patterning of polydimethylsiloxane (PDMS) into complex two-dimensional (2D) or 3D shapes is a crucial step for diverse applications based on soft lithography. Nevertheless, mould replication that incorporates time-consuming and costly photolithography processes still remains the dominant technology in the field. Here we developed monolithic quasi-3D digital patterning of PDMS using laser pyrolysis. In contrast with conventional burning or laser ablation of transparent PDMS, which yields poor surface properties, our successive laser pyrolysis technique converts PDMS into easily removable silicon carbide via consecutive photothermal pyrolysis guided by a continuous-wave laser. We obtained high-quality 2D or 3D PDMS structures with complex patterning starting from a PDMS monolith in a remarkably low prototyping time (less than one hour). Moreover, we developed distinct microfluidic devices with elaborated channel architectures and a customizable organ-on-a-chip device using this approach, which showcases the potential of the successive laser pyrolysis technique for the fabrication of devices for several technological applications.


Asunto(s)
Dimetilpolisiloxanos/química , Dispositivos Laboratorio en un Chip , Rayos Láser , Nylons/química , Pirólisis
4.
Biotechnol Bioeng ; 119(2): 566-574, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34716703

RESUMEN

In vitro platforms for studying the human brain have been developed, and brain organoids derived from stem cells have been studied. However, current organoid models lack three-dimensional (3D) vascular networks, limiting organoid proliferation, differentiation, and apoptosis. In this study, we created a 3D model of vascularized spheroid cells using an injection-molded microfluidic chip. We cocultured spheroids derived from induced neural stem cells (iNSCs) with perfusable blood vessels. Gene expression analysis and immunostaining revealed that the vascular network greatly enhanced spheroid differentiation and reduced apoptosis. This platform can be used to further study the functional and structural interactions between blood vessels and neural spheroids, and ultimately to simulate brain development and disease.


Asunto(s)
Técnicas de Cocultivo/métodos , Dispositivos Laboratorio en un Chip , Neovascularización Fisiológica/fisiología , Células-Madre Neurales/citología , Esferoides Celulares/citología , Apoptosis/fisiología , Vasos Sanguíneos/fisiología , Diferenciación Celular/fisiología , Humanos , Ingeniería de Tejidos
5.
Biotechnol Bioeng ; 119(12): 3678-3693, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36043394

RESUMEN

The development of a scalable and highly reproducible in vitro tumor microenvironment (TME) platform still sheds light on new insights into cancer metastasis mechanisms and anticancer therapeutic strategies. Here, we present an all-in-one injection molded plastic array three-dimensional culture platform (All-in-One-IMPACT) that integrates vascularized tumor spheroids for highly reproducible, high-throughput experimentation. This device allows the formation of self-assembled cell spheroids on a chip by applying the hanging drop method to the cell culture channel. Then, when the hydrogel containing endothelial cells and fibroblasts is injected, the spheroid inside the droplet can be patterned together in three dimensions along the culture channel. In just two steps above, we can build a vascularized TME within a defined area. This process does not require specialized user skill and minimizes error-inducing steps, enabling both reproducibility and high throughput of the experiment. We have successfully demonstrated the process, from spheroid formation to tumor vascularization, using patient-derived cancer cells (PDCs) as well as various cancer cell lines. Furthermore, we performed combination therapies with Taxol (paclitaxel) and Avastin (bevacizumab), which are used in standard care for metastatic cancer. The All-in-One IMPACT is a powerful tool for establishing various anticancer treatment strategies through the development of a complex TME for use in high-throughput experiments.


Asunto(s)
Microfluídica , Neoplasias , Humanos , Células Endoteliales , Reproducibilidad de los Resultados , Esferoides Celulares , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
6.
Adv Exp Med Biol ; 1379: 231-256, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35760994

RESUMEN

Tumors disrupt the normal homeostasis of human body as they proliferate in abnormal speed. For constant proliferation, tumors recruit new blood vessels transporting nutrients and oxygen. Immune system simultaneously recruits lymphatic vessels to induce the death of tumor cells. Hence, understanding tumor dynamics are important to developing anti-cancer therapies. Tumor-on-a-chip technology can be applied to identify the structural and functional units of tumors and tumor microenvironments with high reproducibility and reliability, monitoring the development and pathophysiology of tumors, and predicting drug effectiveness. Herein, we explore the ability of tumor-on-a-chip technology to mimic angiogenic and lymphangiogenic tumor microenvironments of organs. Microfluidic systems allow elaborate manipulation of the development and status of cancer. Therefore, they can be used to validate the effects of various drug combinations, specify them, and assess the factors that influence cancer treatment. We discuss the mechanisms of action of several drugs for cancer treatment in terms of tumor growth and progression involving angiogenesis and lymphangiogenesis. Moreover, we present future applications of emerging tumor-on-a-chip technology for drug development and cancer therapy.


Asunto(s)
Dispositivos Laboratorio en un Chip , Neoplasias , Humanos , Microfluídica , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Reproducibilidad de los Resultados , Microambiente Tumoral
7.
Proc Natl Acad Sci U S A ; 116(24): 12007-12012, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31118285

RESUMEN

Fragile X syndrome (FXS) is the leading monogenic cause of autism and intellectual disability. FXS is caused by loss of expression of fragile X mental retardation protein (FMRP), an RNA-binding protein that regulates translation of numerous mRNA targets, some of which are present at synapses. While protein synthesis deficits have long been postulated as an etiology of FXS, how FMRP loss affects distributions of newly synthesized proteins is unknown. Here we investigated the role of FMRP in regulating expression of new copies of the synaptic protein PSD95 in an in vitro model of synaptic plasticity. We find that local BDNF application promotes persistent accumulation of new PSD95 at stimulated synapses and dendrites of cultured neurons, and that this accumulation is absent in FMRP-deficient mouse neurons. New PSD95 accumulation at sites of BDNF stimulation does not require known mechanisms regulating FMRP-mRNA interactions but instead requires the PI3K-mTORC1-S6K1 pathway. Surprisingly, in FMRP-deficient neurons, BDNF induction of new PSD95 accumulation can be restored by mTORC1-S6K1 blockade, suggesting that constitutively high mTORC1-S6K1 activity occludes PSD95 regulation by BDNF and that alternative pathways exist to mediate induction when mTORC1-S6K1 is inhibited. This study provides direct evidence for deficits in local protein synthesis and accumulation of newly synthesized protein in response to local stimulation in FXS, and supports mTORC1-S6K1 pathway inhibition as a potential therapeutic approach for FXS.


Asunto(s)
Homólogo 4 de la Proteína Discs Large/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Neuronas/metabolismo , Animales , Dendritas/metabolismo , Síndrome del Cromosoma X Frágil/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Endogámicos C57BL , Plasticidad Neuronal/fisiología , Ratas , Transducción de Señal/fisiología , Sinapsis/metabolismo
8.
Hum Reprod ; 36(10): 2720-2731, 2021 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-34363466

RESUMEN

STUDY QUESTION: Can we reconstitute physiologically relevant 3-dimensional (3D) microengineered endometrium in-vitro model? SUMMARY ANSWER: Our representative microengineered vascularised endometrium on-a-chip closely recapitulates the endometrial microenvironment that consists of three distinct layers including epithelial cells, stromal fibroblasts and endothelial cells in a 3D extracellular matrix in a spatiotemporal manner. WHAT IS KNOWN ALREADY: Organ-on-a-chip, a multi-channel 3D microfluidic cell culture system, is widely used to investigate physiologically relevant responses of organ systems. STUDY DESIGN, SIZE, DURATION: The device consists of five microchannels that are arrayed in parallel and partitioned by array of micropost. Two central channels are for 3D culture and morphogenesis of stromal fibroblast and endothelial cells. In addition, the outermost channel is for the culture of additional endometrial stromal fibroblasts that secrete biochemical cues to induce directional pro-angiogenic responses of endothelial cells. To seed endometrial epithelial cells, on Day 8, Ishikawa cells were introduced to one of the two medium channels to adhere on the gel surface. After that, the microengineered endometrium was cultured for an additional 5-6 days (total ∼ 14 days) for the purpose of each experiment. PARTICIPANTS/MATERIALS, SETTING, METHODS: Microfluidic 3D cultures were maintained in endothelial growth Medium 2 with or without oestradiol and progesterone. Some cultures additionally received exogenous pro-angiogenic factors. For the three distinct layers of microengineered endometrium-on-a-chip, the epithelium, stroma and blood vessel characteristics and drug response of each distinct layer in the microfluidic model were assessed morphologically and biochemically. The quantitative measurement of endometrial drug delivery was evaluated by the permeability coefficients. MAIN RESULTS AND THE ROLE OF CHANCE: We established microengineered vascularised endometrium-on-chip, which consists of three distinct layers: epithelium, stroma and blood vessels. Our endometrium model faithfully recapitulates in-vivo endometrial vasculo-angiogenesis and hormonal responses displaying key features of the proliferative and secretory phases of the menstrual cycle. Furthermore, the effect of the emergency contraception drug levonorgestrel was evaluated in our model demonstrating increased endometrial permeability and blood vessel regression in a dose-dependent manner. We finally provided a proof of concept of the multi-layered endometrium model for embryo implantation, which aids a better understanding of the molecular and cellular mechanisms underlying this process. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: This report is largely an in-vitro study and it would be beneficial to validate our findings using human primary endometrial cells. WIDER IMPLICATIONS OF THE FINDINGS: Our 3D microengineered vascularised endometrium-on-a-chip provides a new in-vitro approach to drug screening and drug discovery by mimicking the complicated behaviours of human endometrium. Thus, we suggest our model as a tool for addressing critical challenges and unsolved problems in female diseases, such as endometriosis, uterine cancer and female infertility, in a personalised manner. STUDY FUNDING/COMPETING INTEREST(S): This work is supported by funding from the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) to Y.J.K. (No. 2018R1C1B6003), to J.A. (No. 2020R1I1A1A01074136) and to H.S.K. (No. 2020R1C1C100787212). The authors report no conflicts of interest.


Asunto(s)
Células Endoteliales , Dispositivos Laboratorio en un Chip , Implantación del Embrión , Endometrio , Femenino , Humanos , Ciclo Menstrual
9.
Biotechnol Bioeng ; 118(7): 2524-2535, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33764506

RESUMEN

In vitro models are becoming more advanced to truly present physiological systems while enabling high-throughput screening and analysis. Organ-on-a-chip devices provide remarkable results through the reconstruction of a three-dimensional (3D) cellular microenvironment although they need to be further developed in terms of multiple liquid patterning principle, material properties, and scalability. Here we present a 3D anchor-based microfluidic injection-molded plastic array culture platform (Anchor-IMPACT) that enables selective, space-intensive patterning of hydrogels using anchor-island for high-throughput angiogenesis evaluation model. Anchor-IMPACT consists of a central channel and an anchor-island, integrating the array into an abbreviated 96-well plate format with a standard microscope slide size. The anchor-island enables selective 3D cell patterning without channel-to-channel contact or any hydrogel injection port using an anchor structure unlike conventional culture compartment. The hydrogel was patterned into defined regions by spontaneous capillary flow under hydrophilic conditions. We configured multiple cell patterning structures to investigate the angiogenic potency of colorectal cancer cells in Anchor-IMPACT and the morphological properties of the angiogenesis induced by the paracrine effect were evaluated. In addition, the efficacy of anticancer drugs against angiogenic sprouts was verified by following dose-dependent responses. Our results indicate that Anchor-IMPACT offers not only a model of high-throughput experimentation but also an advanced 3D cell culture platform and can significantly improve current in vitro models while providing the basis for developing predictive preclinical models for biopharmaceutical applications.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Dispositivos Laboratorio en un Chip , Técnicas Analíticas Microfluídicas , Neovascularización Patológica/tratamiento farmacológico , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Humanos , Neovascularización Patológica/metabolismo
10.
Int J Mol Sci ; 22(19)2021 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-34639099

RESUMEN

Renal fibrosis is a progressive chronic kidney disease that ultimately leads to end-stage renal failure. Despite several approaches to combat renal fibrosis, an experimental model to evaluate currently available drugs is not ideal. We developed fibrosis-mimicking models using three-dimensional (3D) co-culture devices designed with three separate layers of tubule interstitium, namely, epithelial, fibroblastic, and endothelial layers. We introduced human renal proximal tubular epithelial cells (HK-2), human umbilical-vein endothelial cells, and patient-derived renal fibroblasts, and evaluated the effects of transforming growth factor-ß (TGF-ß) and TGF-ß inhibitor treatment on this renal fibrosis model. The expression of the fibrosis marker alpha smooth muscle actin upon TGF-ß1 treatment was augmented in monolayer-cultured HK-2 cells in a 3D disease model. In the vascular compartment of renal fibrosis models, the density of vessels was increased and decreased in the TGF-ß-treated group and TGF-ß-inhibitor treatment group, respectively. Multiplex ELISA using supernatants in the TGF-ß-stimulating 3D models showed that pro-inflammatory cytokine and growth factor levels including interleukin-1 beta, tumor necrosis factor alpha, basic fibroblast growth factor, and TGF-ß1, TGF-ß2, and TGF-ß3 were increased, which mimicked the fibrotic microenvironments of human kidneys. This study may enable the construction of a human renal fibrosis-mimicking device model beyond traditional culture experiments.


Asunto(s)
Endotelio Vascular/patología , Fibroblastos/patología , Fibrosis/patología , Túbulos Renales Proximales/patología , Impresión Tridimensional/instrumentación , Factor de Crecimiento Transformador beta1/farmacología , Células Cultivadas , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibrosis/inducido químicamente , Fibrosis/metabolismo , Humanos , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo
11.
Mol Syst Biol ; 15(11): e8947, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31777174

RESUMEN

Stimulation of PC-12 cells with epidermal (EGF) versus nerve (NGF) growth factors (GFs) biases the distribution between transient and sustained single-cell ERK activity states, and between proliferation and differentiation fates within a cell population. We report that fibroblast GF (FGF2) evokes a distinct behavior that consists of a gradually changing population distribution of transient/sustained ERK signaling states in response to increasing inputs in a dose response. Temporally controlled GF perturbations of MAPK signaling dynamics applied using microfluidics reveal that this wider mix of ERK states emerges through the combination of an intracellular feedback, and competition of FGF2 binding to FGF receptors (FGFRs) and heparan sulfate proteoglycan (HSPG) co-receptors. We show that the latter experimental modality is instructive for model selection using a Bayesian parameter inference. Our results provide novel insights into how different receptor tyrosine kinase (RTK) systems differentially wire the MAPK network to fine-tune fate decisions at the cell population level.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Animales , Teorema de Bayes , Relación Dosis-Respuesta a Droga , Proteoglicanos de Heparán Sulfato/metabolismo , Técnicas Analíticas Microfluídicas , Células PC12 , Ratas , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo
12.
Biotechnol Bioeng ; 117(3): 748-762, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31709508

RESUMEN

The human central nervous system (CNS) vasculature expresses a distinctive barrier phenotype, the blood-brain barrier (BBB). As the BBB contributes to low efficiency in CNS pharmacotherapy by restricting drug transport, the development of an in vitro human BBB model has been in demand. Here, we present a microfluidic model of CNS angiogenesis having three-dimensional (3D) lumenized vasculature in concert with perivascular cells. We confirmed the necessity of the angiogenic tri-culture system (brain endothelium in direct interaction with pericytes and astrocytes) to attain essential phenotypes of BBB vasculature, such as minimized vessel diameter and maximized junction expression. In addition, lower vascular permeability is achieved in the tri-culture condition compared to the monoculture condition. Notably, we focussed on reconstituting the functional efflux transporter system, including p-glycoprotein (p-gp), which is highly responsible for restrictive drug transport. By conducting the calcein-AM efflux assay on our 3D perfusable vasculature after treatment of efflux transporter inhibitors, we confirmed the higher efflux property and prominent effect of inhibitors in the tri-culture model. Taken together, we designed a 3D human BBB model with functional barrier properties based on a developmentally inspired CNS angiogenesis protocol. We expect the model to contribute to a deeper understanding of pathological CNS angiogenesis and the development of effective CNS medications.


Asunto(s)
Transporte Biológico/fisiología , Barrera Hematoencefálica , Dispositivos Laboratorio en un Chip , Técnicas Analíticas Microfluídicas/instrumentación , Modelos Biológicos , Neovascularización Fisiológica/fisiología , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/fisiología , Células Cultivadas , Diseño de Equipo , Humanos , Pericitos/citología
13.
Proc Natl Acad Sci U S A ; 114(51): 13471-13476, 2017 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-29196524

RESUMEN

Cells experience compressive stress while growing in limited space or migrating through narrow constrictions. To survive such stress, cells reprogram their intracellular organization to acquire appropriate mechanical properties. However, the mechanosensors and downstream signaling networks mediating these changes remain largely unknown. Here, we have established a microfluidic platform to specifically trigger compressive stress, and to quantitatively monitor single-cell responses of budding yeast in situ. We found that yeast senses compressive stress via the cell surface protein Mid2 and the calcium channel proteins Mid1 and Cch1, which then activate the Pkc1/Mpk1 MAP kinase pathway and calcium signaling, respectively. Genetic analysis revealed that these pathways work in parallel to mediate cell survival. Mid2 contains a short intracellular tail and a serine-threonine-rich extracellular domain with spring-like properties, and both domains are required for mechanosignaling. Mid2-dependent spatial activation of the Pkc1/Mpk1 pathway depolarizes the actin cytoskeleton in budding or shmooing cells, thereby antagonizing polarized growth to protect cells under compressive stress conditions. Together, these results identify a conserved signaling network responding to compressive mechanical stress, which, in higher eukaryotes, may ensure cell survival in confined environments.


Asunto(s)
Calcineurina/metabolismo , Canales de Calcio/metabolismo , Proteína Quinasa C/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Estrés Mecánico , Citoesqueleto de Actina/metabolismo , Calcineurina/genética , Canales de Calcio/genética , Señalización del Calcio , Supervivencia Celular , Pared Celular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Sistema de Señalización de MAP Quinasas , Mecanotransducción Celular , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Microfluídica/instrumentación , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteína Quinasa C/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética
14.
Biotechnol Bioeng ; 116(10): 2425-2438, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31180148

RESUMEN

Axonal regeneration and remyelination of peripheral motor neurons (MNs) are critical for restoring neuromuscular motor function after injury or peripheral neuropathy. We examined whether optogenetically mediated light stimulation (OMLS) could enhance the axon outgrowth and myelination of MNs using three-dimensional motor neuron-Schwann cell (MN-SC) coculture on a microfluidic biochip. The biochip was designed to allow SCs to interact with the axons of MNs, while preventing direct contact between SCs and the cell bodies of MNs. Following coculture with SCs on the microfluidic biochip, MNs were transfected with a light-sensitive channelrhodopsin gene. Transfected MNs subjected to repeated light stimulation (20 Hz, 1 hr) produced significantly longer axons than nontransfected MNs. OMLS of MNs greatly increased the number of myelin basic protein (MBP)-expressing SCs, promoting the initiation of myelination of MNs. Ultrastructurally, OMLS of MNs markedly enhanced the thickness of the compact myelin sheath around the MN axons such that the average thickness was closer to that of the theoretical estimates in vivo. Thus, the MN-SC coculture model on a microfluidic biochip augmented by OMLS of MNs is a feasible platform for studying the relationship of neuronal activity with regrowth and remyelination.


Asunto(s)
Axones/metabolismo , Dispositivos Laboratorio en un Chip , Neuronas Motoras/metabolismo , Optogenética , Células de Schwann/metabolismo , Animales , Técnicas de Cocultivo , Ratones , Neuronas Motoras/citología , Células de Schwann/citología
15.
Nephrol Dial Transplant ; 34(1): 49-62, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29726981

RESUMEN

Background: Our laboratory has previously demonstrated that Sirt1endo-/- mice show endothelial dysfunction and exaggerated renal fibrosis, whereas mice with silenced endothelial transforming growth factor beta (TGF-ß) signaling are resistant to fibrogenic signals. Considering the fact that the only difference between these mutant mice is confined to the vascular endothelium, this indicates that secreted substances contribute to these contrasting responses. Methods: We performed an unbiased proteomic analysis of the secretome of renal microvascular endothelial cells (RMVECs) isolated from these two mutants. We cultured renal fibroblasts and RMVECs and used microfluidic devices for coculturing. Results: Dickkopf-3 (DKK3), a putative ligand of the Wnt/ß-catenin pathway, was present exclusively in the fibrogenic secretome. In cultured fibroblasts, DKK3 potently induced myofibroblast activation. In addition, DKK3 antagonized effects of DKK1, a known inhibitor of the Wnt pathway, in conversion of fibroblasts to myofibroblasts. In RMVECs, DKK3 induced endothelial-mesenchymal transition and impaired their angiogenic competence. The inhibition of endothelial outgrowth, enhanced myofibroblast formation and endothelial-mesenchymal transition were confirmed in coculture. In reporter DKK3-eGFP × Col3.6-GFPcyan mice, DKK3 was marginally expressed under basal conditions. Adriamycin-induced nephropathy resulted in upregulation of DKK3 expression in tubular and, to a lesser degree, endothelial compartments. Sulindac sulfide was found to exhibit superior Wnt pathway-suppressive action and decreased DKK3 signals and the extent of renal fibrosis. Conclusions: In conclusion, this unbiased proteomic screen of the profibrogenic endothelial secretome revealed DKK3 acting as an agonist of the Wnt pathway, enhancing formation of myofibroblasts and endothelial-mesenchymal transition and impairing angiogenesis. A potent inhibitor of the Wnt pathway, sulindac sulfide, suppressed nephropathy-induced DKK3 expression and renal fibrosis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Endotelio Vascular/patología , Transición Epitelial-Mesenquimal , Fibrosis/patología , Enfermedades Renales/patología , Proteoma/análisis , Receptor Tipo II de Factor de Crecimiento Transformador beta/fisiología , Sirtuina 1/fisiología , Animales , Endotelio Vascular/metabolismo , Fibrosis/metabolismo , Enfermedades Renales/metabolismo , Ratones , Ratones Noqueados , Proteómica , Vía de Señalización Wnt , beta Catenina/metabolismo
16.
J Liposome Res ; 29(1): 44-52, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29262741

RESUMEN

Resistance to chemotherapy is a key factor in the inefficacy of various forms of treatments for cancer. In the present study, chemo-resistant proteins, including glucose-regulated protein 78 (GRP78)/clusterin (CLU) targeted 1,2-dioleoyloxy-3-trimethylammoniumpropane (DOTAP) liposomes, were developed as a delivery system for co-delivery of camptothecin (CPT) and GRP78 siRNA/CLU siRNA. Their drug/gene co-deliveries were quantitatively assessed in cancer stem cells (CSC) and MCF-7 cells. DOTAP-CPT/siRNA were prepared via electrostatic interaction on GRP78 siRNA or CLU siRNA. The size and ζ-potential of liposomes and lipoplexes were measured by dynamic light scattering techniques and electrophoretic light scattering spectrophotometry. The lipoplexes formation was tested by using gel electrophoresis. Immunofluorescence analysis showed that the expression level of CLU and GRP78 were significantly elevated in CSC compared to MCF-7 cells. Transfection and drug-delivery efficiency of DOTAP-CPT/siRNA were quantitatively compared with Lipofectamine 2000. Compared to free CPT, DOTAP-CPT-siCLU delivery in CSC and MCF-7 cells increased transfection efficiency and chemo-sensitivity by 4.1- and 5.9-fold, respectively. On the other hand, DOTAP-CPT-siGRP78 delivery increased transfection efficiency and chemo sensitivity by 4.4- and 6.2-fold in CSC and MCF-7 cells, respectively, compared to free CPT. It is significant that 3 ± 1.2-fold increase in transfection efficiency was achieved by lipofectamine. Consequently, an increase in anti-cancer/gene silencing efficacy was quantitatively observed as an effect of DOTAP-CPT/siRNA treatment, which was relatively higher than lipofectamine treatment. Conclusively, our experimental data quantitatively demonstrate that using DOTAP-CPT-siRNA specifically targeting (CSCs) chemo-resistant protein in vitro offers substantial potential for synergistic anti-cancer therapy.


Asunto(s)
Antineoplásicos Fitogénicos , Camptotecina , Clusterina/antagonistas & inhibidores , Liposomas , Células Madre Neoplásicas , Antineoplásicos Fitogénicos/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Camptotecina/administración & dosificación , Clusterina/genética , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Chaperón BiP del Retículo Endoplásmico , Femenino , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Técnicas de Transferencia de Gen , Humanos , Liposomas/química , Células MCF-7 , Células Madre Neoplásicas/efectos de los fármacos , ARN Interferente Pequeño/administración & dosificación
17.
Mikrochim Acta ; 186(4): 211, 2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30830307

RESUMEN

A graphene oxide (GO)-based cost-effective, automatted strip test has developed for screening of inhibitors of endonuclease EcoRV. The method involves the use of GO and a DNA substrate for EcoRV that contains both an ssDNA region for binding of GO and a fluorescein amidite (FAM)-labelled dsDNA. All the components were inkjet printed on a piece of parchment paper. The ssDNA region binds to the surface of GO and anchors so that the fluorescence of FAM is quenched. The parchment paper strip is then incubated with a sample containing EcoRV which causes enzymatic hydrolysis, and dsDNA was separated from the GO. As a result, green fluorescence is generated at the reaction spot. Enzyme activity can be measured in the presence and absence of aurintricarboxy acid acting as an EcoRV inhibitor. This method excels by its need for 2-3 orders less reagents compared to the standard well plate assay. Thus, it is an efficient platform for GO-based screening of EcoRV enzyme inhibitors. Graphical abstract A graphene oxide (GO)-based endonuclease EcoRV inhibition FRET assay using inkjet printing was developed. Printing of GO along with assay reagents has a beneficial effect on the enzymatic reaction on paper. This method was successfully applied to evaluate EcoRV inhibitor activity.


Asunto(s)
ADN/química , Inhibidores Enzimáticos/química , Exonucleasas/antagonistas & inhibidores , Fluoresceína/química , Grafito/química , Secuencia de Bases , Bioensayo/métodos , Transferencia Resonante de Energía de Fluorescencia/métodos , Colorantes Fluorescentes/química , Impresión Tridimensional , Espectrometría de Fluorescencia/métodos
18.
Chemphyschem ; 19(10): 1123-1127, 2018 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-29542276

RESUMEN

The schwann cells of the peripheral nervous system are indispensable for the formation, maintenance, and modulation of synapses over the life cycle. They not only recognize neuron-glia signaling molecules, but also secrete gliotransmitters. Through these processes, they regulate neuronal excitability and thus the release of neurotransmitters from the nerve terminal at the neuromuscular junction. Gliotransmitters strongly affect nerve communication, and their secretion is mainly triggered by synchronized Ca2+ signaling, implicating Ca2+ waves in synapse function. Reciprocally, neurotransmitters released during synaptic activity can evoke increases in intracellular Ca2+ levels. A reconsideration of the interplay between the two main types of cells in the nervous system is due, as the concept of nervous system activity comprising only neuron-neuron and neuron-muscle action has become untenable. A more precise understanding of the roles of schwann cells in nerve-muscle signaling is required.


Asunto(s)
Células de Schwann/metabolismo , Sinapsis/metabolismo , Animales , Humanos , Células de Schwann/citología
19.
Langmuir ; 34(1): 173-178, 2018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29198119

RESUMEN

Fluorescent Au nanocrystals (AuNCs)-silica hybrid nanocomposite (FLASH) was synthesized by co-condensation of surface-modified AuNCs. Present FLASH nanocomposite exhibited four times the enhanced photoluminescence and photocatalytic activity compared to single nanocrystals. On the basis of these enhanced optical features, we successfully demonstrated in vitro fluorescence bioimaging of introduced FLASH to human cervical cancer cell line (HeLa). Beyond the confirmation of photocatalytic activity from the photodegradation of methylene blue as a model compound, the regional selective photodynamic therapy of HeLa cells under UV irradiation was also presented. Taken together the enhanced optical features and further potential in theranostic applications, we expect that the present FLASH can be a promising tool for nanobiotechnology field.


Asunto(s)
Colorantes Fluorescentes/química , Oro/química , Nanopartículas del Metal/química , Nanocompuestos/química , Fármacos Fotosensibilizantes/química , Dióxido de Silicio/química , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/efectos de la radiación , Bencimidazoles/química , Catálisis , Fluorescencia , Colorantes Fluorescentes/metabolismo , Colorantes Fluorescentes/farmacología , Colorantes Fluorescentes/efectos de la radiación , Glutatión/química , Células HeLa , Humanos , Nanopartículas del Metal/efectos de la radiación , Azul de Metileno/química , Nanocompuestos/efectos de la radiación , Fármacos Fotosensibilizantes/metabolismo , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/efectos de la radiación , Silanos/química , Dióxido de Silicio/síntesis química , Dióxido de Silicio/farmacología , Dióxido de Silicio/efectos de la radiación , Nanomedicina Teranóstica , Rayos Ultravioleta
20.
Mol Syst Biol ; 11(11): 838, 2015 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-26613961

RESUMEN

Transient versus sustained ERK MAP kinase (MAPK) activation dynamics induce proliferation versus differentiation in response to epidermal (EGF) or nerve (NGF) growth factors in PC-12 cells. Duration of ERK activation has therefore been proposed to specify cell fate decisions. Using a biosensor to measure ERK activation dynamics in single living cells reveals that sustained EGF/NGF application leads to a heterogeneous mix of transient and sustained ERK activation dynamics in distinct cells of the population, different than the population average. EGF biases toward transient, while NGF biases toward sustained ERK activation responses. In contrast, pulsed growth factor application can repeatedly and homogeneously trigger ERK activity transients across the cell population. These datasets enable mathematical modeling to reveal salient features inherent to the MAPK network. Ultimately, this predicts pulsed growth factor stimulation regimes that can bypass the typical feedback activation to rewire the system toward cell differentiation irrespective of growth factor identity.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Biología de Sistemas/métodos , Animales , Técnicas Biosensibles , Transferencia Resonante de Energía de Fluorescencia , Técnicas Analíticas Microfluídicas , Modelos Biológicos , Células PC12 , Ratas , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA