Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
J Cell Mol Med ; 22(5): 2896-2907, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29516686

RESUMEN

Midazolam, a benzodiazepine derivative, is widely used for sedation and surgery. However, previous studies have demonstrated that Midazolam is associated with increased risks of congenital malformations, such as dwarfism, when used during early pregnancy. Recent studies have also demonstrated that Midazolam suppresses osteogenesis of mesenchymal stem cells (MSCs). Given that hypertrophic chondrocytes can differentiate into osteoblast and osteocytes and contribute to endochondral bone formation, the effect of Midazolam on chondrogenesis remains unclear. In this study, we applied a human MSC line, the KP cell, to serve as an in vitro model to study the effect of Midazolam on chondrogenesis. We first successfully established an in vitro chondrogenic model in a micromass culture or a 2D high-density culture performed with TGF-ß-driven chondrogenic induction medium. Treatment of the Midazolam dose-dependently inhibited chondrogenesis, examined using Alcian blue-stained glycosaminoglycans and the expression of chondrogenic markers, such as SOX9 and type II collagen. Inhibition of Midazolam by peripheral benzodiazepine receptor (PBR) antagonist PK11195 or small interfering RNA rescued the inhibitory effects of Midazolam on chondrogenesis. In addition, Midazolam suppressed transforming growth factor-ß-induced Smad3 phosphorylation, and this inhibitory effect could be rescued using PBR antagonist PK11195. This study provides a possible explanation for Midazolam-induced congenital malformations of the musculoskeletal system through PBR.


Asunto(s)
Condrogénesis/efectos de los fármacos , Antagonistas de Receptores de GABA-A/farmacología , Células Madre Mesenquimatosas/metabolismo , Midazolam/farmacología , Receptores de GABA-A/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Humanos , Isoquinolinas/farmacología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
2.
Biochem Biophys Res Commun ; 497(3): 869-875, 2018 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-29470990

RESUMEN

Cell migration is a critical process during development, tissue repair, and cancer metastasis. It requires complex processes of cell adhesion, cytoskeletal dynamics, and force generation. Lis1 plays an important role in the migration of neurons, fibroblasts and other cell types, and is essential for normal development of the cerebral cortex. Mutations in human LIS1 gene cause classical lissencephaly (smooth brain), resulting from defects in neuronal migration. However, how Lis1 may affect force generation in migrating cells is still not fully understood. Using traction force microscopy (TFM) with live cell imaging to measure cellular traction force in migrating NIH3T3 cells, we showed that Lis1 knockdown (KD) by RNA interference (RNAi) caused reductions in cell migration and traction force against the extracellular matrix (ECM). Immunostaining of cytoskeletal components in Lis1 KD cells showed disorganization of microtubules and actin filaments. Interestingly, focal adhesions at the cell periphery were significantly reduced. These results suggest that Lis1 is important for cellular traction force generation through the regulation of cytoskeleton organization and focal adhesion formation in migrating cells.


Asunto(s)
1-Alquil-2-acetilglicerofosfocolina Esterasa/metabolismo , Movimiento Celular , Citoesqueleto/metabolismo , Fibroblastos/citología , Proteínas Asociadas a Microtúbulos/metabolismo , 1-Alquil-2-acetilglicerofosfocolina Esterasa/genética , Animales , Fenómenos Biomecánicos , Fibroblastos/metabolismo , Adhesiones Focales/metabolismo , Ratones , Proteínas Asociadas a Microtúbulos/genética , Células 3T3 NIH , Interferencia de ARN
3.
Cells Tissues Organs ; 204(3-4): 179-190, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28848167

RESUMEN

Recently, a novel substrate known as an electrospun polylactic acid (PLLA) microtube array membrane (MTAM) was successfully developed as a cell coculture platform. Structurally, this substrate is made up of one-to-one connected, ultrathin, submicron scale fibers that are arranged in an arrayed formation. Its unique structure confers several key advantages which are beneficial in a cell coculture system. In this study, the interaction between rat fetal neural stem cells (NSC) and astrocytes was examined by comparing the outcome of a typical Transwell-based coculture system and that of an electrospun PLLA MTAM-based coculture system. Compared to tissue culture polystyrene (TCP) and Transwell coculture inserts, a superior cell viability of NSC was observed when cultured in lumens of electrospun PLLA MTAM (with supportive immunostaining images). Reverse transcription polymerase chain reaction revealed a strong interaction between astrocytes and NSC through a higher expression of doublecortin and a lower expression of nestin. These data demonstrate that MTAM is clearly a better coculture platform than the traditional Transwell system.


Asunto(s)
Astrocitos/química , Regeneración Nerviosa/fisiología , Ingeniería de Tejidos/métodos , Animales , Técnicas de Cocultivo , Proteína Doblecortina , Humanos , Ratas
4.
J Cell Sci ; 127(Pt 19): 4186-200, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25107365

RESUMEN

Focal adhesions (FAs) undergo maturation that culminates in size and composition changes that modulate adhesion, cytoskeleton remodeling and differentiation. Although it is well recognized that stimuli for osteogenesis of mesenchymal stem cells (MSCs) drive FA maturation, actin organization and stress fiber polarization, the extent to which FA-mediated signals regulated by the FA protein composition specifies MSC commitment remains largely unknown. Here, we demonstrate that, upon dexamethasone (osteogenic induction) treatment, guanine nucleotide exchange factor H1 (GEF-H1, also known as Rho guanine nucleotide exchange factor 2, encoded by ARHGEF2) is significantly enriched in FAs. Perturbation of GEF-H1 inhibits FA formation, anisotropic stress fiber orientation and MSC osteogenesis in an actomyosin-contractility-independent manner. To determine the role of GEF-H1 in MSC osteogenesis, we explore the GEF-H1-modulated FA proteome that reveals non-muscle myosin-II heavy chain-B (NMIIB, also known as myosin-10, encoded by MYH10) as a target of GEF-H1 in FAs. Inhibition of targeting NMIIB into FAs suppresses FA formation, stress fiber polarization, cell stiffness and osteogenic commitments in MSCs. Our data demonstrate a role for FA signaling in specifying MSC commitment.


Asunto(s)
Adhesiones Focales/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Diferenciación Celular/fisiología , Linaje de la Célula , Humanos , Osteogénesis , Transducción de Señal
5.
J Cell Mol Med ; 19(5): 934-47, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25683605

RESUMEN

Cancer metastasis occurs via a progress involving abnormal cell migration. Cell migration, a dynamic physical process, is controlled by the cytoskeletal system, which includes the dynamics of actin organization and cellular adhesive organelles, focal adhesions (FAs). However, it is not known whether the organization of actin cytoskeletal system has a regulatory role in the physiologically relevant aspects of cancer metastasis. In the present studies, it was found that lung adenocarcinoma cells isolated from the secondary lung cancer of the lymph nodes, H1299 cells, show specific dynamics in terms of the actin cytoskeleton and FAs. This results in a higher level of mobility and this is regulated by an immature FA component, ß-PIX (PAK-interacting exchange factor-ß). In H1299 cells, ß-PIX's activity was found not to be down-regulated by sequestration onto stress fibres, as the cells did not bundle actin filaments into stress fibres. Thus, ß-PIX mainly remained localized at FAs, which allowed maturation of nascent adhesions into focal complexes; this resulted in actin polymerization, increased actin network integrity, changes in the intracellular microrheology at the peripheral of the cell, and cell polarity, which in turn regulated cell migration. Perturbation of ß-PIX caused an inhibition of cell migration, including migration velocity, accumulated distance and directional persistence. Our results demonstrate the importance of ß-PIX to the regulation of high mobility of lung adenocarcinoma cell line H1299 and that this occurs via regulation of FA dynamics, changes in actin cytoskeleton organization and cell polarity.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Movimiento Celular , Citoplasma/metabolismo , Adhesiones Focales/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Línea Celular Tumoral , Polaridad Celular , Regulación hacia Abajo , Elasticidad , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Microscopía Confocal , Miosina Tipo II/metabolismo , Interferencia de ARN , Factores de Intercambio de Guanina Nucleótido Rho/genética , Fibras de Estrés/metabolismo , Imagen de Lapso de Tiempo/métodos , Viscosidad
6.
J Cell Sci ; 126(Pt 5): 1260-7, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23378028

RESUMEN

Stromal interaction molecule 1 (STIM1) is an endoplasmic reticulum (ER) Ca(2+) sensor that triggers the store-operated Ca(2+) entry (SOCE). The clinical relevance of STIM1 has been highlighted in breast and cervical cancer, but the molecular mechanism by which STIM1 promotes cancer progression remains unclear. This study explores the regulatory mechanisms by which STIM1-dependent Ca(2+) signaling controls cancer cell migration. Three different SOCE inhibitors, SKF96365, 2-APB and YM-58483, significantly inhibited cervical cancer cell migration to a similar extent to that of STIM1 silencing. In contrast, STIM1 overexpression significantly enhanced cervical cancer cell migration. Live cell confocal images and three-dimensional tomograms showed that STIM1 formed aggregates and translocated towards the plasma membranes of migratory cells, and this was accompanied by increasing cytosolic Ca(2+) spikes. STIM1 silencing also inhibited the recruitment and association of active focal adhesion kinase (pTyr397-FAK) and talin at focal adhesions, indicating the blockade of force transduction from integrin signaling. Epidermal growth factor-induced phosphorylation of myosin II regulatory light chains was abolished by STIM1 knockdown and SOCE inhibition. Dual immunostaining of activated myosin II (pSer19-MLC) and actin revealed that actomyosin formation depended on STIM1-mediated Ca(2+) entry. Most importantly, STIM1 expression levels as well as SOCE activity controlled the generation of cell contractile force, as measured by the microfabricated post-array-detector system. These results highlight the unique role of STIM1-dependent Ca(2+) signaling in controlling cell migration by the regulation of actomyosin reorganization in conjunction with enhanced contractile forces.


Asunto(s)
Actomiosina/metabolismo , Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Anilidas/farmacología , Compuestos de Boro/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Humanos , Imidazoles/farmacología , Immunoblotting , Microscopía Confocal , Interferencia de ARN , Molécula de Interacción Estromal 1 , Tiadiazoles/farmacología
7.
Exp Dermatol ; 24(8): 579-84, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25877039

RESUMEN

One of the key features of keloid is its fibroblasts migrating beyond the original wound border. During migration, cells not only undergo molecular changes but also mechanical modulation. This process is led by actin filaments serving as the backbone of intra-cellular force and transduces external mechanical signal via focal adhesion complex into the cell. Here, we focus on determining the mechanical changes of actin filaments and the spatial distribution of forces in response to changing chemical stimulations and during cell migration. Atomic force microscopy and micropost array detector are used to determine and compare the magnitude and distribution of filament elasticity and force generation in fibroblasts and keloid fibroblasts. We found both filament elasticity and force generation show spatial distribution in a polarized and migrating cell. Such spatial distribution is disrupted when mechano-signalling is perturbed by focal adhesion kinase inhibitor and in keloid fibroblasts. The demonstration of keloid pathology at the nanoscale highlights the coupling of cytoskeletal function with physical characters at the subcellular level and provides new research directions for migration-related disease such as keloid.


Asunto(s)
Citoesqueleto/fisiología , Fibroblastos/fisiología , Queloide/patología , Citoesqueleto de Actina/fisiología , Animales , Movimiento Celular , Polaridad Celular , Elasticidad , Fibroblastos/ultraestructura , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/fisiología , Adhesiones Focales/fisiología , Humanos , Ratones , Microscopía de Fuerza Atómica , Células 3T3 NIH , Quinolonas/farmacología , Estrés Mecánico , Sulfonas/farmacología , Cicatrización de Heridas
8.
J BUON ; 19(1): 137-44, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24659655

RESUMEN

PURPOSE: Irradiation-induced autophagy has been reported in several types of cancers, however, the relationship between irradiation and autophagy in human oral squamous cell carcinoma (OSCC) has not yet been described. In this study we investigated the induction of autophagy in cell lines by exposing them to ionizing irradiation. METHODS: Human OSCC OC3 and SAS cell lines were used in this study. Cell viability and induction of autophagy were determined under irradiation treatment. The GFP-LC3 puncta formation and the levels of LC3-II as indicators of autophagy were detected by fluorescence microscopy and Western blot method. The signaling pathways involved in irradiation-mediated autophagy were also determined by Western blot method. RESULTS: Irradiation decreased cell viability only in OC3 cells, while autophagic machinery and related signaling pathways were found to be elevated after irradiation in OC3 and SAS cells. However, autophagic degradation determined by the reduction of p62 levels was only found in OC3 cells, suggesting autophagosome accumulation took place in SAS cells. In addition, irradiation accompanied with rapamycin treatment elevated autophagy formation and induced death of OC3 cells. CONCLUSIONS: These results suggested that induction of autophagy might provide an advantageous strategy to increase the anticancer effects of radiotherapy in patients with OSCCs.


Asunto(s)
Autofagia/efectos de la radiación , Carcinoma de Células Escamosas/radioterapia , Neoplasias de la Boca/radioterapia , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Apoptosis/efectos de la radiación , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral/efectos de la radiación , Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Radiación Ionizante , Proteína Sequestosoma-1 , Transducción de Señal/efectos de la radiación
9.
J Cell Mol Med ; 17(7): 823-32, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23672518

RESUMEN

Stem cells have been shown to have the potential to provide a source of cells for applications to tissue engineering and organ repair. The mechanisms that regulate stem cell fate, however, mostly remain unclear. Mesenchymal stem cells (MSCs) are multipotent progenitor cells that are isolated from bone marrow and other adult tissues, and can be differentiated into multiple cell lineages, such as bone, cartilage, fat, muscles and neurons. Although previous studies have focused intensively on the effects of chemical signals that regulate MSC commitment, the effects of physical/mechanical cues of the microenvironment on MSC fate determination have long been neglected. However, several studies provided evidence that mechanical signals, both direct and indirect, played important roles in regulating a stem cell fate. In this review, we summarize a number of recent studies on how cell adhesion and mechanical cues influence the differentiation of MSCs into specific lineages. Understanding how chemical and mechanical cues in the microenvironment orchestrate stem cell differentiation may provide new insights into ways to improve our techniques in cell therapy and organ repair.


Asunto(s)
Diferenciación Celular , Células Madre Mesenquimatosas/citología , Animales , Adhesión Celular , Linaje de la Célula , Fuerza Compresiva , Citoesqueleto/fisiología , Humanos , Transducción de Señal , Estrés Mecánico , Distribución Tisular , Ingeniería de Tejidos/métodos
10.
Nat Methods ; 7(9): 733-6, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20676108

RESUMEN

We report the establishment of a library of micromolded elastomeric micropost arrays to modulate substrate rigidity independently of effects on adhesive and other material surface properties. We demonstrated that micropost rigidity impacts cell morphology, focal adhesions, cytoskeletal contractility and stem cell differentiation. Furthermore, early changes in cytoskeletal contractility predicted later stem cell fate decisions in single cells.


Asunto(s)
Células Endoteliales/citología , Células Madre Mesenquimatosas/citología , Polímeros/química , Estrés Mecánico , Adhesión Celular/fisiología , Células Cultivadas , Dimetilpolisiloxanos/química , Elastómeros , Humanos , Ensayo de Materiales , Microscopía de Fuerza Atómica/instrumentación , Tamaño de la Partícula , Silicio/química , Propiedades de Superficie
11.
Sci Technol Adv Mater ; 14(5): 054401, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27877605

RESUMEN

Stem cells are known for their potential to repair damaged tissues. The adhesion, growth and differentiation of stem cells are likely controlled by the surrounding microenvironment which contains both chemical and physical cues. Physical cues in the microenvironment, for example, nanotopography, were shown to play important roles in stem cell fate decisions. Thus, controlling stem cell behavior by nanoscale topography has become an important issue in stem cell biology. Nanotechnology has emerged as a new exciting field and research from this field has greatly advanced. Nanotechnology allows the manipulation of sophisticated surfaces/scaffolds which can mimic the cellular environment for regulating cellular behaviors. Thus, we summarize recent studies on nanotechnology with applications to stem cell biology, including the regulation of stem cell adhesion, growth, differentiation, tracking and imaging. Understanding the interactions of nanomaterials with stem cells may provide the knowledge to apply to cell-scaffold combinations in tissue engineering and regenerative medicine.

12.
J Cell Commun Signal ; 17(2): 361-370, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37191841

RESUMEN

Shear stress is a frictional drag generated by the flow of fluid, such as blood or interstitial fluid, and plays a critical role in regulating cellular gene expression and functional phenotype. The matricellular CCN family proteins are dynamically regulated by shear stress of different flow patterns, and their expression significantly alters the microenvironment of cells. Secreted CCN proteins mainly bind to several cell surface integrin receptors to mediate their diverse functions in regulating cell survival, function, and behavior. Gene-knockout studies indicate major functions of CCN proteins in the cardiovascular and skeletal systems, the two primary systems in which CCN expressions are regulated by shear stress. In the cardiovascular system, the endothelium is directly exposed to vascular shear stress. Unidirectional laminar blood flow generates laminar shear stress, which promotes a mature endothelial phenotype and upregulates anti-inflammatory CCN3 expression. In contrast, disturbed flow generates oscillatory shear stress, which induces endothelial dysfunction through the induction of CCN1 and CCN2. Shear-induced CCN1 binds to integrin α6ß1 and promotes superoxide production, NF-κB activation, and inflammatory gene expression in endothelial cells. Although the interaction between shear stress and CCN4-6 is not clear, CCN 4 exhibits a proinflammatory property and CCN5 inhibits vascular cell growth and migration. The crucial roles of CCN proteins in cardiovascular development, homeostasis, and disease are evident but not fully understood. In the skeletal system, mechanical loading on bone generates shear stress from interstitial fluid in the lacuna-canalicular system and promotes osteoblast differentiation and bone formation. CCN1 and CCN2 are induced and potentially mediate fluid shear stress mechanosensing in osteocytes. However, the exact roles of interstitial shear stress-induced CCN1 and CCN2 in bone are still not clear. In contrast to other CCN family proteins, CCN3 inhibits osteoblast differentiation, although its regulation by interstitial shear stress in osteocytes has not been reported. The induction of CCN proteins by shear stress in bone and their functions remain largely unknown and merit further investigation. This review discusses the expression and functions of CCN proteins regulated by shear stress in physiological conditions, diseases, and cell culture models. The roles between CCN family proteins can be compensatory or counteractive in tissue remodeling and homeostasis.

13.
Acta Biomater ; 163: 287-301, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36328121

RESUMEN

Within the heterogeneous tissue architecture, a comprehensive understanding of how cell shapes regulate cytoskeletal mechanics by adjusting focal adhesions (FAs) signals to correlate with the lineage commitment of mesenchymal stromal cells (MSCs) remains obscure. Here, via engineered extracellular matrices, we observed that the development of mature FAs, coupled with a symmetrical pattern of radial fiber bundles, appeared at the right-angle vertices in cells with square shape. While circular cells aligned the transverse fibers parallel to the cell edge, and moved them centripetally in a counter-clockwise direction, symmetrical bundles of radial fibers at the vertices of square cells disrupted the counter-clockwise swirling and bridged the transverse fibers to move centripetally. In square cells, the contractile force, generated by the myosin IIA-enriched transverse fibers, were concentrated and transmitted outwards along the symmetrical bundles of radial fibers, to the extracellular matrix through FAs, and thereby driving FA organization and maturation. The symmetrical radial fiber bundles concentrated the transverse fibers contractility inward to the linkage between the actin cytoskeleton and the nuclear envelope. The tauter cytoskeletal network adjusted the nuclear-actomyosin force balance to cause nuclear deformability and to increase nuclear translocation of the transcription co-activator YAP, which in turn modulated the switch in MSC commitment. Thus, FAs dynamically respond to geometric cues and remodel actin cytoskeletal network to re-distribute intracelluar tension towards the cell nucleus, and thereby controlling YAP mechanotransduction signaling in regulating MSC fate decision. STATEMENT OF SIGNIFICANCE: We decipher how cellular mechanics is self-organized depending on extracellular geometric features to correlate with mesenchymal stromal cell lineage commitment. In response to geometry constrains on cell morphology, symmetrical radial fiber bundles are assembled and clustered depending on the maturation state of focal adhesions and bridge with the transverse fibers, and thereby establishing the dynamic cytoskeletal network. Contractile force, generated by the myosin-IIA-enriched transverse fibers, is transmitted and dynamically drives the retrograde movement of the actin cytoskeletal network, which appropriately adjusts the nuclear-actomyosin force balance and deforms the cell nucleus for YAP mechano-transduction signaling in regulating mesenchymal stromal cell fate decision.


Asunto(s)
Actinas , Células Madre Mesenquimatosas , Actinas/metabolismo , Actomiosina/metabolismo , Mecanotransducción Celular , Forma de la Célula , Osteogénesis , Diferenciación Celular , Factores de Transcripción/metabolismo
14.
Polymers (Basel) ; 14(9)2022 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-35567092

RESUMEN

BACKGROUND: In vitro three-dimensional (3D) hepatic spheroid culture has shown great promise in toxicity testing because it better mimics the cell-cell and cell-matrix interactions found in in vivo conditions than that of the traditional two-dimensional (2D) culture. Despite embedding HepaRG spheroids with collagen type I (collagen I) extracellular matrix (ECM) revealed a much better differentiation capability, almost all the collagen utilized in in vitro hepatocytes cultures is animal-derived collagen that may limit its use in human toxicity testing. METHOD: Here, a preliminary investigation of HepaRG cells cultured in different dimensionalities and with the addition of ECM was performed. Comparisons of conventional 2D culture with 3D spheroid culture were performed based on their functional or structural differences over 7 days. Rat tail collagen (rtCollagen) I and recombinant human collagen (rhCollagen) I were investigated for their ability in promoting HepaRG spheroid differentiation. RESULTS: An immunofluorescence analysis of the hepatocyte-specific functional protein albumin suggested that HepaRG spheroids demonstrated better hepatic function than spheroids from 2D culture, and the function of HepaRG spheroids improved in a time-dependent manner. The fluorescence intensities per unit area of spheroids formed by 1000 cells on days 7 and 10 were 25.41 and 45.38, respectively, whereas almost undetectable fluorescence was obtained with 2D cells. In addition, the embedding of HepaRG spheroids into rtCollagen and rhCollagen I showed that HepaRG differentiation can be accelerated relative to the differentiation of spheroids grown in suspension, demonstrating the great promise of HepaRG spheroids. CONCLUSIONS: The culture conditions established in this study provide a potentially novel alternative for promoting the differentiation of HepaRG spheroids into mature hepatocytes through a collagen-embedded in vitro liver spheroid model. This culture method is envisioned to provide insights for future drug toxicology.

15.
Front Bioeng Biotechnol ; 10: 877480, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35586553

RESUMEN

Blood vessels are ubiquitous in the human body and play essential roles not only in the delivery of vital oxygen and nutrients but also in many disease implications and drug transportation. Although fabricating in vitro blood vessels has been greatly facilitated through various microfluidic organ-on-chip systems, most platforms that are used in the laboratories suffer from a series of laborious processes ranging from chip fabrication, optimization, and control of physiologic flows in micro-channels. These issues have thus limited the implementation of the technique to broader scientific communities that are not ready to fabricate microfluidic systems in-house. Therefore, we aimed to identify a commercially available microfluidic solution that supports user custom protocol developed for microvasculature-on-a-chip (MVOC). The custom protocol was validated to reliably form a smooth and functional blood vessel using a viscous fingering (VF) technique. Using VF technique, the unpolymerized collagen gel in the media channels was extruded by less viscous fluid through VF passive flow pumping, whereby the fluid volume at the inlet and outlet ports are different. The different diameters of hollow tubes produced by VF technique were carefully investigated by varying the ambient temperature, the pressure of the passive pump, the pre-polymerization time, and the concentration of collagen type I. Subsequently, culturing human umbilical vein endothelial cells inside the hollow structure to form blood vessels validated that the VF-created structure revealed a much greater permeability reduction than the vessel formed without VF patterns, highlighting that a more functional vessel tube can be formed in the proposed methodology. We believe the current protocol is timely and will offer new opportunities in the field of in vitro MVOC.

16.
Biomedicines ; 10(7)2022 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-35884894

RESUMEN

Cancer is one of the leading causes of death worldwide. There are only limited treatment strategies that can be applied to treat cancer, including surgical resection, chemotherapy, and radiotherapy, but these have only limited effectiveness. Developing a new drug for cancer therapy is protracted, costly, and inefficient. Recently, drug repurposing has become a rising research field to provide new meaning for an old drug. By searching a drug repurposing database ReDO_DB, a brief list of anesthetic/sedative drugs, such as haloperidol, ketamine, lidocaine, midazolam, propofol, and valproic acid, are shown to possess anti-cancer properties. Therefore, in the current review, we will provide a general overview of the anti-cancer mechanisms of these anesthetic/sedative drugs and explore the potential underlying signaling pathways and clinical application of these drugs applied individually or in combination with other anti-cancer agents.

17.
Int J Oncol ; 60(2)2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35029282

RESUMEN

For a number of years, oral cancer has remained in the top ten most common types of cancer, with an incidence rate that is steadily increasing. In total, ~75% oral cancer cases are associated with lifestyle factors, including uncontrolled alcohol consumption, betel and tobacco chewing, and the excessive use of tobacco. Notably, betel chewing is highly associated with oral cancer in Southeast Asia. Arsenic is a key environmental toxicant; however, arsenic trioxide has been used as a medicine for the treatment of acute promyelocytic leukemia, highlighting its anticancer properties. The present study aimed to investigate the role of arsenic compounds in the treatment of cancer, using FaDu oral squamous carcinoma cells treated with sodium arsenite (NaAsO2) and dimethyl arsenic acid (DMA). The results demonstrated that FaDu cells exhibited membrane blebbing phenomena and high levels of apoptosis following treatment with 10 µM NaAsO2 and 1 mM DMA for 24 h. The results of cell viability assay demonstrated that the rate of FaDu cell survival was markedly reduced as the concentration of arsenic compounds increased from 10 to 100 µM NaAsO2, and 1 to 100 mM DMA. Moreover, flow cytometry was carried out to further examine the effects of arsenic compounds on FaDu cell cycle regulation; the results revealed that treatment with NaAsO2 and DMA led to a significant increase in the percentage of FaDu cells in the sub­G1 and G2/M phases of the cell cycle. An Annexin V/PI double staining assay was subsequently performed to verify the levels of FaDu cell apoptosis following treatment with arsenic compounds. Furthermore, the results of the western blot analyses revealed that the expression levels of caspase­8, ­9 and ­3, and poly ADP­ribose polymerase, as well the levels of phosphorylated JNK and ERK1/2 were increased following treatment with NaAsO2 and DMA in the FaDu cells. On the whole, the results of the present study revealed that treatment with NaAsO2 and DMA promoted the apoptosis of FaDu oral cancer cells, by activating MAPK pathways, as well as the extrinsic and intrinsic apoptotic pathways.


Asunto(s)
Apoptosis/efectos de los fármacos , Arsénico/farmacología , Transducción de Señal/efectos de los fármacos , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Arsénico/metabolismo , Caspasas/metabolismo , Caspasas/farmacología , Supervivencia Celular/efectos de los fármacos , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/farmacología , Carcinoma de Células Escamosas de Cabeza y Cuello/fisiopatología
18.
Am J Pathol ; 177(4): 1743-54, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20709799

RESUMEN

Transforming growth factor-ß1 (TGF-ß1)-induced epithelial-to-mesenchymal transition (EMT) contributes to the pathophysiological development of kidney fibrosis. Although it was reported that TGF-ß1 enhances ß(1) integrin levels in NMuMG cells, the detailed molecular mechanisms underlying TGF-ß1-induced ß(1) integrin gene expression and the role of ß(1) integrin during EMT in the renal system are still unclear. In this study, we examined the role of ß(1) integrin in TGF-ß1-induced EMT both in vitro and in vivo. TGF-ß1-induced augmentation of ß(1) integrin expression was required for EMT in several epithelial cell lines, and knockdown of Smad3 inhibited TGF-ß1-induced augmentation of ß(1) integrin. TGF-ß1 triggered ß(1) integrin gene promoter activity as assessed by luciferase activity assay. Both knockdown of Smad3 and mutation of the Smad-binding element to block binding to the ß(1) integrin promoter markedly reduced TGF-ß1-induced ß(1) integrin promoter activity. Chromatin immunoprecipitation assay showed that TGF-ß1 enhanced Smad3 binding to the ß(1) integrin promoter. Furthermore, induction of unilateral ureteral obstruction triggered increases of ß(1) integrin in both renal epithelial and interstitial cells. In human kidney with chronic tubulointerstitial fibrosis, we also found a concomitant increase of ß(1) integrin and α-smooth muscle actin in tubule epithelia. Blockade of ß(1) integrin signaling dampened the progression of fibrosis. Taken together, ß(1) integrin mediates EMT and subsequent tubulointerstitutial fibrosis, suggesting that inhibition of ß(1) integrin is a possible therapeutic target for prevention of renal fibrosis.


Asunto(s)
Diferenciación Celular , Transición Epitelial-Mesenquimal/fisiología , Integrina beta1/genética , Nefritis Intersticial/fisiopatología , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Obstrucción Ureteral/patología , Animales , Apoptosis , Western Blotting , Adhesión Celular , Proliferación Celular , Inmunoprecipitación de Cromatina , Enfermedad Crónica , Perros , Femenino , Regulación de la Expresión Génica , Humanos , Técnicas para Inmunoenzimas , Integrina beta1/metabolismo , Queratinocitos/citología , Queratinocitos/metabolismo , Riñón/citología , Riñón/metabolismo , Células LLC-PK1 , Luciferasas/metabolismo , Masculino , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Proteína smad3/genética , Porcinos , Factor de Crecimiento Transformador beta1/genética , Obstrucción Ureteral/metabolismo
19.
Life (Basel) ; 11(12)2021 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-34947927

RESUMEN

Despite improvements in cancer treatments resulting in higher survival rates, the proliferation and metastasis of tumors still raise new questions in cancer therapy. Therefore, new drugs and strategies are still needed. Midazolam (MDZ) is a common sedative drug acting through the γ-aminobutyric acid receptor in the central nervous system and also binds to the peripheral benzodiazepine receptor (PBR) in peripheral tissues. Previous studies have shown that MDZ inhibits cancer cell proliferation but increases cancer cell apoptosis through different mechanisms. In this study, we investigated the possible anticancer mechanisms of MDZ on different cancer cell types. MDZ inhibited transforming growth factor ß (TGF-ß)-induced cancer cell proliferation of both A549 and MCF-7 cells. MDZ also inhibited TGF-ß-induced cell migration, invasion, epithelial-mesenchymal-transition, and Smad phosphorylation in both cancer cell lines. Inhibition of PBR by PK11195 rescued the MDZ-inhibited cell proliferation, suggesting that MDZ worked through PBR to inhibit TGF-ß pathway. Furthermore, MDZ inhibited proliferation, migration, invasion and levels of mesenchymal proteins in MDA-MD-231 triple-negative breast cancer cells. Together, MDZ inhibits cancer cell proliferation both in epithelial and mesenchymal types and EMT, indicating an important role for MDZ as a candidate to treat lung and breast cancers.

20.
Cell Death Discov ; 7(1): 35, 2021 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-33597503

RESUMEN

During differentiation, skeletal muscle develops mature multinucleated muscle fibers, which could contract to exert force on a substrate. Muscle dysfunction occurs progressively in patients with muscular dystrophy, leading to a loss of the ability to walk and eventually to death. The synthetic glucocorticoid dexamethasone (Dex) has been used therapeutically to treat muscular dystrophy by an inhibition of inflammation, followed by slowing muscle degeneration and stabilizing muscle strength. Here, in mice with muscle injury, we found that Dex significantly promotes muscle regeneration via promoting kinesin-1 motor activity. Nevertheless, how Dex promotes myogenesis through kinesin-1 motors remains unclear. We found that Dex directly increases kinesin-1 motor activity, which is required for the expression of a myogenic marker (muscle myosin heavy chain 1/2), and also for the process of myoblast fusion and the formation of polarized myotubes. Upon differentiation, kinesin-1 mediates the recruitment of integrin ß1 onto microtubules allowing delivery of the protein into focal adhesions. Integrin ß1-mediated focal adhesion signaling then guides myoblast fusion towards a polarized morphology. By imposing geometric constrains via micropatterns, we have proved that cell adhesion is able to rescue the defects caused by kinesin-1 inhibition during the process of myogenesis. These discoveries reveal a mechanism by which Dex is able to promote myogenesis, and lead us towards approaches that are more efficient in improving skeletal muscle regeneration.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA