Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Int J Mol Sci ; 25(5)2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38473701

RESUMEN

This work analyzes the role of the tight junction (TJ) protein ZO-2 on mechanosensation. We found that the lack of ZO-2 reduced apical membrane rigidity measured with atomic force microscopy, inhibited the association of γ-actin and JAM-A to the cell border, and instead facilitated p114RhoGEF and afadin accumulation at the junction, leading to an enhanced mechanical tension at the TJ measured by FRET, with a ZO-1 tension probe, and increased tricellular TJ tension. Simultaneously, adherens junction tension measured with an E-cadherin probe was unaltered. The stability of JAM-A and ZO-2 binding was assessed by a collaborative in silico study. The absence of ZO-2 also impacted the cell response to the substrate, as monolayers plated in 20 kPa hydrogels developed holes not seen in parental cultures and displayed a retarded elongation and formation of cell aggregates. The absence of ZO-2 was sufficient to induce YAP and Snail nuclear accumulation in cells cultured over glass, but when ZO-2 KD cells were plated in nanostructured ridge arrays, they displayed an increased abundance of nuclear Snail and conspicuous internalization of claudin-4. These results indicate that the absence of ZO-2 also impairs the response of cells to substrate stiffness and exacerbates transformation triggered by substrate topography.


Asunto(s)
Actinas , Uniones Estrechas , Actinas/metabolismo , Uniones Estrechas/metabolismo , Proteína de la Zonula Occludens-1/metabolismo , Fosfoproteínas/metabolismo
2.
Biol Cell ; 112(10): 265-279, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32449790

RESUMEN

BACKGROUND INFORMATION: There have been several studies to understand the influence of stiffness of the culture substrates for different types of adherent cells. It is generally accepted that cell proliferation, spreading and focal adhesions increase with higher matrix stiffness. However, what remains unclear is whether this kind of cell behaviour may be reverted by culturing on soft substrates those cell lines that were originally selected or primed on stiff surfaces. RESULTS: Here, we studied the influence of substrate softness on proliferation, adhesion and morphology of the highly proliferative hepatic C9 cell line. We cultured C9 cells on soft and stiff polydimethylsiloxane (PDMS) substrates prepared with two different elastic moduli in the range of 10 and 200 kPa, respectively. Lower cell proliferation was observed on substrates with lower stiffness without affecting cell viability. The proliferation rate of C9 cell line along with extracellular growth-regulated kinase (ERK) phosphorylation was decreased on soft PDMS. Despite this cell line has been described as a hepatic epithelial cell, our characterisation demonstrated that the origin of C9 cells is uncertain, although clearly epithelial, with the expression of markers of several hepatic cells. Surprisingly, consecutive passages of C9 cells on soft PDMS did not alter this mesenchymal phenotype, vimentin expression did not decrease when culturing cells in soft substrates, even though the ERK phosphorylation levels eventually were increased after several passages on soft PDMS, triggering again an increase of cell proliferation. CONCLUSIONS AND SIGNIFICANCE: This study shows that the exposure of C9 cells to soft substrates promoted a decrease of cell proliferation rate, as reported for other types of cells on PDMS, whereas a much longer term exposure caused cells to adapt to softness after trained for several passages, reactivating proliferation. During this phenomenon, the morphology and phenotype of trained cells was modified accompanying the increase of cell proliferation rate contrary to the effect observed in short periods of cell culture. In contrast to previous reports, cell death was not observed during these experiments, discarding a cell selection mechanism and suggesting soft cell adaptation may be limited in time in C9 cells.


Asunto(s)
Medios de Cultivo/química , Células Epiteliales/citología , Hepatocitos/citología , Biomarcadores/metabolismo , Adhesión Celular , Línea Celular , Proliferación Celular , Dimetilpolisiloxanos/química , Humanos
3.
Eur J Immunol ; 47(8): 1305-1316, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28586109

RESUMEN

Transforming growth factor-ß (TGF-ß) is a potent mast cell (MC) chemoattractant able to modulate local inflammatory reactions. The molecular mechanism leading to TGF-ß-directed MC migration is not fully described. Here we analyzed the role of the Src family protein kinase Fyn on the main TGF-ß-induced cytoskeletal changes leading to MC migration. Utilizing bone marrow-derived mast cells (BMMCs) from WT and Fyn-deficient mice we found that BMMC migration to TGF-ß was impaired in the absence of the kinase. TGF-ß caused depolymerization of the cortical actin ring and changes on the phosphorylation of cofilin, LIMK and CAMKII only in WT cells. Defective cofilin activation and phosphorylation of regulatory proteins was detected in Fyn-deficient BMMCs and this finding correlated with a lower activity of the catalytic subunit of the phosphatase PP2A. Diminished TGF-ß-induced chemotaxis of Fyn-deficient cells was also observed in an in vivo model of MC migration (bleomycin-induced scleroderma). Our results show that Fyn kinase is an important positive effector of TGF-ß-induced chemotaxis through the control of PP2A activity and this is relevant to pathological processes that are related to TGF-ß-dependent mast cell migration.


Asunto(s)
Actinas/metabolismo , Quimiotaxis , Mastocitos/fisiología , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Factores Despolimerizantes de la Actina/metabolismo , Animales , Mastocitos/inmunología , Ratones , Ácido Ocadaico/farmacología , Fosforilación , Proteína Fosfatasa 2/metabolismo , Proteínas Proto-Oncogénicas c-fyn/genética , Transducción de Señal , Proteína Smad2/metabolismo
4.
J Immunol ; 196(12): 5075-88, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27183589

RESUMEN

Mast cells produce proinflammatory cytokines in response to TLR4 ligands, but the signaling pathways involved are not fully described. In this study, the participation of the Src family kinase Fyn in the production of TNF after stimulation with LPS was evaluated using bone marrow-derived mast cells from wild-type and Fyn-deficient mice. Fyn(-/-) cells showed higher LPS-induced secretion of preformed and de novo-synthesized TNF. In both cell types, TNF colocalized with vesicle-associated membrane protein (VAMP)3-positive compartments. Addition of LPS provoked coalescence of VAMP3 and its interaction with synaptosomal-associated protein 23; those events were increased in the absence of Fyn. Higher TNF mRNA levels were also observed in Fyn-deficient cells as a result of increased transcription and greater mRNA stability after LPS treatment. Fyn(-/-) cells also showed higher LPS-induced activation of TAK-1 and ERK1/2, whereas IκB kinase and IκB were phosphorylated, even in basal conditions. Increased responsiveness in Fyn(-/-) cells was associated with a lower activity of protein phosphatase 2A (PP2A) and augmented activity of protein kinase C (PKC)α/ß, which was dissociated from PP2A and increased its association with the adapter protein neuroblast differentiation-associated protein (AHNAK, desmoyokin). LPS-induced PKCα/ß activity was associated with VAMP3 coalescence in WT and Fyn-deficient cells. Reconstitution of MC-deficient Wsh mice with Fyn(-/-) MCs produced greater LPS-dependent production of TNF in the peritoneal cavity. Our data show that Fyn kinase is activated after TLR4 triggering and exerts an important negative control on LPS-dependent TNF production in MCs controlling the inactivation of PP2Ac and activation of PKCα/ß necessary for the secretion of TNF by VAMP3(+) carriers.


Asunto(s)
Regulación de la Expresión Génica , Mastocitos/inmunología , Proteína Quinasa C-alfa/metabolismo , Proteína Fosfatasa 2/metabolismo , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Receptor Toll-Like 4/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Lipopolisacáridos/inmunología , Mastocitos/efectos de los fármacos , Ratones , Fosforilación , Unión Proteica , Proteínas Proto-Oncogénicas c-fyn/deficiencia , Proteínas Proto-Oncogénicas c-fyn/genética , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Proteína 3 de Membrana Asociada a Vesículas/metabolismo
5.
J Biol Chem ; 290(7): 4487-99, 2015 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-25561741

RESUMEN

TGF-ß-induced antimitotic signals are highly regulated during cell proliferation under normal and pathological conditions, such as liver regeneration and cancer. Up-regulation of the transcriptional cofactors Ski and SnoN during liver regeneration may favor hepatocyte proliferation by inhibiting TGF-ß signals. In this study, we found a novel mechanism that regulates Ski protein stability through TGF-ß and G protein-coupled receptor (GPCR) signaling. Ski protein is distributed between the nucleus and cytoplasm of normal hepatocytes, and the molecular mechanisms controlling Ski protein stability involve the participation of actin cytoskeleton dynamics. Cytoplasmic Ski is partially associated with actin and localized in cholesterol-rich vesicles. Ski protein stability is decreased by TGF-ß/Smads, GPCR/Rho signals, and actin polymerization, whereas GPCR/cAMP signals and actin depolymerization promote Ski protein stability. In conclusion, TGF-ß and GPCR signals differentially regulate Ski protein stability and sorting in hepatocytes, and this cross-talk may occur during liver regeneration.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Proteínas de Unión al ADN/metabolismo , Endosomas/metabolismo , Hepatocitos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Western Blotting , Núcleo Celular/metabolismo , Proliferación Celular , Células Cultivadas , Citoplasma/metabolismo , Técnica del Anticuerpo Fluorescente , Células Hep G2 , Hepatocitos/citología , Humanos , Inmunoprecipitación , Regeneración Hepática , Masculino , Ratas , Ratas Wistar , Transducción de Señal
6.
Biochim Biophys Acta ; 1850(9): 1832-41, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26002202

RESUMEN

BACKGROUND: Ski and SnoN proteins function as transcriptional co-repressors in the TGF-ß pathway. They regulate cell proliferation and differentiation, and their aberrant expression results in altered TGF-ß signalling, malignant transformation, and alterations in cell proliferation. METHODS: We carried out a comparative characterization of the endogenous Ski and SnoN protein regulation by TGF-ß, cell adhesion disruption and actin-cytoskeleton rearrangements between normal and transformed hepatocytes; we also analyzed Ski and SnoN protein stability, subcellular localization, and how their protein levels impact the TGF-ß/Smad-driven gene transcription. RESULTS: Ski and SnoN protein levels are lower in normal hepatocytes than in hepatoma cells. They exhibit a very short half-life and a nuclear/cytoplasmic distribution in normal hepatocytes opposed to a high stability and restricted nuclear localization in hepatoma cells. Interestingly, while normal cells exhibit a transient TGF-ß-induced gene expression, the hepatoma cells are characterized by a strong and sustained TGF-ß-induced gene expression. A novel finding is that Ski and SnoN stability is differentially regulated by cell adhesion and cytoskeleton rearrangements in the normal hepatocytes. The inhibition of protein turnover down-regulated both Ski and SnoN co-repressors impacting the kinetic of expression of TGF-ß-target genes. CONCLUSION: Normal regulatory mechanisms controlling Ski and SnoN stability, subcellular localization and expression are altered in hepatocarcinoma cells. GENERAL SIGNIFICANCE: This work provides evidence that Ski and SnoN protein regulation is far more complex in normal than in transformed cells, since many of the normal regulatory mechanisms are lost in transformed cells.


Asunto(s)
Actinas/química , Citoesqueleto/química , Proteínas de Unión al ADN/química , Hepatocitos/metabolismo , Péptidos y Proteínas de Señalización Intracelular/química , Proteínas Proto-Oncogénicas/química , Animales , Carcinoma Hepatocelular/metabolismo , Adhesión Celular , Línea Celular Tumoral , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/metabolismo , Péptidos y Proteínas de Señalización Intracelular/análisis , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Polimerizacion , Estabilidad Proteica , Proteínas Proto-Oncogénicas/análisis , Proteínas Proto-Oncogénicas/metabolismo , Ratas , Proteínas Smad/fisiología , Factor de Crecimiento Transformador beta/farmacología
7.
Biochim Biophys Acta ; 1830(11): 5049-58, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23872350

RESUMEN

BACKGROUND: SnoN and Ski proteins function as Smad transcriptional corepressors and are implicated in the regulation of diverse cellular processes such as proliferation, differentiation and transformation. Transforming growth factor-ß (TGF-ß) signaling causes SnoN and Ski protein degradation via proteasome with the participation of phosphorylated R-Smad proteins. Intriguingly, the antibiotics anisomycin (ANS) and puromycin (PURO) are also able to downregulate Ski and SnoN proteins via proteasome. METHODS: We explored the effects of ANS and PURO on SnoN protein downregulation when the activity of TGF-ß signaling was inhibited by using different pharmacological and non-pharmacological approaches, either by using specific TßRI inhibitors, overexpressing the inhibitory Smad7 protein, or knocking-down TßRI receptor or Smad2 by specific shRNAs. The outcome of SnoN and Ski downregulation induced by ANS or PURO on TGF-ß signaling was also studied. RESULTS: SnoN protein downregulation induced by ANS and PURO did not involve the induction of R-Smad phosphorylation but it was abrogated after TGF-ß signaling inhibition; this effect occurred in a cell type-specific manner and independently of protein synthesis inhibition or any other ribotoxic effect. Intriguingly, antibiotics seem to require components of the TGF-ß/Smad pathway to downregulate SnoN. In addition, SnoN protein downregulation induced by antibiotics favored gene transcription induced by TGF-ß signaling. CONCLUSIONS: ANS and PURO require TGF-ß/Smad pathway to induce SnoN and Ski protein downregulation independently of inducing R-Smad2 phosphorylation, which facilitates TGF-ß signaling. GENERAL SIGNIFICANCE: Antibiotic analogs lacking ribotoxic effects are useful as pharmacological tools to study TGF-ß signaling by controlling Ski and SnoN protein levels.


Asunto(s)
Anisomicina/farmacología , Proteínas Oncogénicas/metabolismo , Puromicina/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Células HeLa , Células Hep G2 , Humanos , Visón/genética , Proteínas Oncogénicas/genética , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína smad7/genética , Proteína smad7/metabolismo , Transcripción Genética/efectos de los fármacos , Factor de Crecimiento Transformador beta1/genética
8.
J Biol Chem ; 287(32): 26764-76, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22674574

RESUMEN

The human SKI-like (SKIL) gene encodes the SMAD transcriptional corepressor SNON that antagonizes TGF-ß signaling. SNON protein levels are tightly regulated by the TGF-ß pathway: whereas a short stimulation with TGF-ß decreases SNON levels by its degradation via the proteasome, longer TGF-ß treatment increases SNON levels by inducing SKIL gene expression. Here, we investigated the molecular mechanisms involved in the self-regulation of SKIL gene expression by SNON. Bioinformatics analysis showed that the human SKIL gene proximal promoter contains a TGF-ß response element (TRE) bearing four groups of SMAD-binding elements that are also conserved in mouse. Two regions of 408 and 648 bp of the human SKIL gene (∼2.4 kb upstream of the ATG initiation codon) containing the core promoter, transcription start site, and the TRE were cloned for functional analysis. Binding of SMAD and SNON proteins to the TRE region of the SKIL gene promoter after TGF-ß treatment was demonstrated by ChIP and sequential ChIP assays. Interestingly, the SNON-SMAD4 complex negatively regulated basal SKIL gene expression through binding the promoter and recruiting histone deacetylases. In response to TGF-ß signal, SNON is removed from the SKIL gene promoter, and then the activated SMAD complexes bind the promoter to induce SKIL gene expression. Subsequently, the up-regulated SNON protein in complex with SMAD4 represses its own expression as part of the negative feedback loop regulating the TGF-ß pathway. Accordingly, when the SNON-SMAD4 complex is absent as in some cancer cells lacking SMAD4 the regulation of some TGF-ß target genes is modified.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteína Smad4/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Humanos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/fisiología , Proteína Smad4/genética , Proteína Smad4/fisiología , Transcripción Genética/fisiología
9.
IUBMB Life ; 65(12): 1035-42, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24273150

RESUMEN

Calcium-sensing receptor (CaSR) contributes to maintain homeostatic levels of extracellular calcium. In addition, CaSR controls other cellular activities such as proliferation and migration, particularly in cells not related to extracellular calcium homeostasis, potentially by cross-talking with parallel signaling pathways. Here we report that CaSR attenuates transforming growth factor-ß (TGF-ß)-signaling in hepatic C9 cells and in transfected HEK293 cells. Wild type CaSR interferes with TGF-ß-dependent Smad2 phosphorylation and induces its proteasomal degradation, resulting in a decrease of TGF-ß-dependent transcriptional activity, whereas an inactivating CaSR mutant does not transduce an inhibitory effect of extracellular calcium on TGF-ß signaling. Attenuation of TGF-ß signaling in response to extracellular calcium is linked to Rab11-dependent CaSR-trafficking with the intervention of CaSR carboxyl-terminal tail. Our data suggest that CaSR might regulate TGF-ß-dependent cellular responses mediated by TGF-ß signaling inhibition.


Asunto(s)
Procesamiento Proteico-Postraduccional , Receptores Sensibles al Calcio/metabolismo , Transducción de Señal , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta1/fisiología , Animales , Células HEK293 , Humanos , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Transporte de Proteínas , Proteolisis , Ratas
10.
Biosensors (Basel) ; 13(8)2023 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-37622879

RESUMEN

In this work, we present an accessible benchtop fabrication technique to obtain a planar array of gold nanowrinkled surface electrodes (ANSE) for the construction of electrochemical cells, specifically to monitor soluble biomarkers of interest in cell culture environments. We present a complete characterization of the array and its response as an electrochemical cell. To validate our sensor, we evaluated the device sensitivity to detect nitric oxide (NO), an important molecule produced by endothelial cells as a response to environmental signals such as mechanics and growth factors. While testing measurements of nitric oxide in aqueous solutions with isotonic salt concentrations, we evidenced the influence of the environmental conditions for such electrochemical measurements, showing that the aqueous medium, usually not accounted for, significantly impacts the outcome. Finally, we present the application of the electrochemical sensor for the detection of nitric oxide released from stimulated endothelial cells as a proof of concept.


Asunto(s)
Células Endoteliales , Óxido Nítrico , Técnicas de Cultivo de Célula , Electrodos , Oro
11.
Heliyon ; 9(11): e21519, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38027697

RESUMEN

The TGF-ß and Hippo pathways are critical for liver size control, regeneration, and cancer progression. The transcriptional cofactor TAZ, also named WWTR1, is a downstream effector of Hippo pathway and plays a key role in the maintenance of liver physiological functions. However, the up-regulation of TAZ expression has been associated with liver cancer progression. Recent evidence shows crosstalk of TGF-ß and Hippo pathways, since TGF-ß modulates TAZ expression through different mechanisms in a cellular context-dependent manner but supposedly independent of SMADs. Here, we evaluate the molecular interplay between TGF-ß pathway and TAZ expression and observe that TGF-ß induces TAZ expression through SMAD canonical pathway in liver cancer HepG2 cells. Therefore, TAZ cofactor is a primary target of TGF-ß/SMAD-signaling, one of the pathways altered in liver cancer.

12.
Methods Mol Biol ; 2174: 277-297, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32813257

RESUMEN

Stiffness control of cell culture platforms provides researchers in cell biology with the ability to study different experimental models in conditions of mimicking physiological or pathological microenvironments. Nevertheless, the signal transduction pathways and drug sensibility of cancer cells have been poorly characterized widely using biomimetic platforms because the limited experience of cancer cell biology groups about handling substrates with specific mechanical properties. The protein cross-linking and stiffening control are crucial checkpoints that could strongly affect cell adhesion and spreading, misrepresenting the data acquired, and also generating inaccurate cellular models. Here, we introduce a simple method to adhere to polyacrylamide (PAA) hydrogels on glass coverslips without any special treatment for mechanics studies in cancer cell biology. By using a commercial photosensitive glue, Loctite 3525, it is possible to polymerize PAA hydrogels directly on glass surfaces. Furthermore, we describe a cross-linking reaction method to attach proteins to PAA as an alternative method to Sulfo-SANPAH cross-linking, which is sometimes difficult to implement and reproduce. In this chapter, we describe a reliable procedure to fabricate ECM protein-cross-linked PAA hydrogels for mechanotransduction studies on cancer cells.


Asunto(s)
Resinas Acrílicas/química , Adhesivos/química , Adhesión Celular , Hidrogeles/química , Neoplasias/patología , Reactivos de Enlaces Cruzados/química , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Proteínas de la Matriz Extracelular/química , Técnica del Anticuerpo Fluorescente , Vidrio , Células Hep G2 , Humanos , Mecanotransducción Celular , Metacrilatos/química , Neoplasias/química , Microambiente Tumoral
13.
Polymers (Basel) ; 13(4)2021 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-33672475

RESUMEN

Following the general aim of recapitulating the native mechanical properties of tissues and organs in vitro, the field of materials science and engineering has benefited from recent progress in developing compliant substrates with physical and chemical properties similar to those of biological materials. In particular, in the field of mechanobiology, soft hydrogels can now reproduce the precise range of stiffnesses of healthy and pathological tissues to study the mechanisms behind cell responses to mechanics. However, it was shown that biological tissues are not only elastic but also relax at different timescales. Cells can, indeed, perceive this dissipation and actually need it because it is a critical signal integrated with other signals to define adhesion, spreading and even more complicated functions. The mechanical characterization of hydrogels used in mechanobiology is, however, commonly limited to the elastic stiffness (Young's modulus) and this value is known to depend greatly on the measurement conditions that are rarely reported in great detail. Here, we report that a simple relaxation test performed under well-defined conditions can provide all the necessary information for characterizing soft materials mechanically, by fitting the dissipation behavior with a generalized Maxwell model (GMM). The simple method was validated using soft polyacrylamide hydrogels and proved to be very useful to readily unveil precise mechanical properties of gels that cells can sense and offer a set of characteristic values that can be compared with what is typically reported from microindentation tests.

14.
Biomimetics (Basel) ; 5(2)2020 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-32630500

RESUMEN

Liver is an essential organ that carries out multiple functions such as glycogen storage, the synthesis of plasma proteins, and the detoxification of xenobiotics. Hepatocytes are the parenchyma that sustain almost all the functions supported by this organ. Hepatocytes and non-parenchymal cells respond to the mechanical alterations that occur in the extracellular matrix (ECM) caused by organogenesis and regenerating processes. Rearrangements of the ECM modify the composition and mechanical properties that result in specific dedifferentiation programs inside the hepatic cells. Quiescent hepatocytes are embedded in the soft ECM, which contains an important concentration of fibrillar collagens in combination with a basement membrane-associated matrix (BM). This work aims to evaluate the role of fibrillar collagens and BM on actin cytoskeleton organization and the function of rat primary hepatocytes cultured on soft elastic polyacrylamide hydrogels (PAA HGs). We used rat tail collagen type I and Matrigel® as references of fibrillar collagens and BM respectively and mixed different percentages of collagen type I in combination with BM. We also used peptides obtained from decellularized liver matrices (dECM). Remarkably, hepatocytes showed a poor adhesion in the absence of collagen on soft PAA HGs. We demonstrated that collagen type I inhibited apoptosis and activated extracellular signal-regulated kinases 1/2 (ERK1/2) in primary hepatocytes cultured on soft hydrogels. Epidermal growth factor (EGF) was not able to rescue cell viability in conjugated BM but affected cell aggregation in soft PAA HGs conjugated with combinations of different proportions of collagen and BM. Interestingly, actin cytoskeleton was localized and preserved close to plasma membrane (cortical actin) and proximal to intercellular ducts (canaliculi-like structures) in soft conditions; however, albumin protein expression was not preserved, even though primary hepatocytes did not remodel their actin cytoskeleton significantly in soft conditions. This investigation highlights the important role of fibrillar collagens on soft hydrogels for the maintenance of survival and aggregation of the hepatocytes. Data suggest evaluating the conditions that allow the establishment of optimal biomimetic environments for physiology and cell biology studies, where the phenotype of primary cells may be preserved for longer periods of time.

15.
Micromachines (Basel) ; 10(9)2019 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-31480301

RESUMEN

Microfluidics has become a very promising technology in recent years, due to its great potential to revolutionize life-science solutions. Generic microfabrication processes have been progressively made available to academic laboratories thanks to cost-effective soft-lithography techniques and enabled important progress in applications like lab-on-chip platforms using rapid- prototyping. However, micron-sized features are required in most designs, especially in biomimetic cell culture platforms, imposing elevated costs of production associated with lithography and limiting the use of such devices. In most cases, however, only a small portion of the structures require high-resolution and cost may be decreased. In this work, we present a replica-molding method separating the fabrication steps of low (macro) and high (micro) resolutions and then merging the two scales in a single chip. The method consists of fabricating the largest possible area in inexpensive macromolds using simple techniques such as plastics micromilling, laser microfabrication, or even by shrinking printed polystyrene sheets. The microfeatures were made on a separated mold or onto existing macromolds using photolithography or 2-photon lithography. By limiting the expensive area to the essential, the time and cost of fabrication can be reduced. Polydimethylsiloxane (PDMS) microfluidic chips were successfully fabricated from the constructed molds and tested to validate our micro-macro method.

16.
ACS Biomater Sci Eng ; 5(9): 4219-4227, 2019 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-33417779

RESUMEN

Polyacrylamide (PAA) hydrogels are now widely used in mechanobiology because the well-defined available protocols allow a robust and reproducible control of substrate stiffness within a physiological range. However, several assays require hydrogels inside traditional plastic substrates and the current methods remain relatively tedious. Here, we present a simple and direct fabrication technique that successfully attaches PAA hydrogels inside polystyrene multiwell plates and Petri dishes of different sizes. It permits a control of the Young's modulus of the gels, within the desired range for mechanobiology. Some critical steps, that had to be overcome to guarantee protein conjugation and cell attachment, are detailed, as they differ from the standardized preparation on glass substrates. To validate our process, we demonstrated that HepG2 and 3T3L1 cell lines as well as primary hepatocytes seeded on PAA gels of different stiffnesses in plastics showed a mechanical response identical to the cells cultured on traditional gels.

17.
Lab Chip ; 19(20): 3512-3525, 2019 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-31544189

RESUMEN

The study of mechanotransduction signals and cell response to mechanical properties requires designing culture substrates that possess some, or ideally all, of the following characteristics: (1) biological compatibility and adhesive properties, (2) stiffness control or tunability in a dynamic mode, (3) patternability on the microscale and (4) integrability in microfluidic chips. The most common materials used to address cell mechanotransduction are hydrogels, due to their softness. However, they may be impractical when complex scaffolds are sought and they lack viscous dissipative properties that are very important in mechanobiology. In this work, we show that an off-the-shelf, biocompatible photosensitive glue, Loctite 3525, may be used readily in mechanobiology assays without any special treatment prior to fabrication of cell culture platforms. Despite a high (MPa) stiffness easily tunable by UV exposure time at a fixed dose, 3T3 fibroblasts showed a response to the mechanics of the material similar to that obtained on much softer (kPa) hydrogels. Loctite's viscous dissipation properties indeed seemed to be responsible for such cell mechanical response, as suggested by recent works where more complex two-phase hydrogels were employed. More interestingly, it was possible to stiffen soft Loctite substrates by post-exposing them during cell culture, to observe changes in cell spreading caused by a dynamic stiffness modification. Thanks to Loctite 3525's patternability, micropillars were also fabricated to demonstrate the compatibility with traction force microscopy studies. Finally, the glue was used as an excellent adhesion layer for hydrogels on glass or PDMS, without the need for additional treatment, enabling the easy fabrication of microfluidic chips integrating hydrogels.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Metacrilatos/química , Microfluídica/instrumentación , Animales , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Técnicas de Cultivo de Célula/instrumentación , Línea Celular , Módulo de Elasticidad , Adhesiones Focales/efectos de los fármacos , Humanos , Hidrogeles/química , Mecanotransducción Celular/fisiología , Metacrilatos/farmacología , Ratones , Rayos Ultravioleta
18.
Artículo en Inglés | MEDLINE | ID: mdl-29892481

RESUMEN

The transforming growth factor-ß (TGF-ß) family plays major pleiotropic roles by regulating many physiological processes in development and tissue homeostasis. The TGF-ß signaling pathway outcome relies on the control of the spatial and temporal expression of >500 genes, which depend on the functions of the Smad protein along with those of diverse modulators of this signaling pathway, such as transcriptional factors and cofactors. Ski (Sloan-Kettering Institute) and SnoN (Ski novel) are Smad-interacting proteins that negatively regulate the TGF-ß signaling pathway by disrupting the formation of R-Smad/Smad4 complexes, as well as by inhibiting Smad association with the p300/CBP coactivators. The Ski and SnoN transcriptional cofactors recruit diverse corepressors and histone deacetylases to repress gene transcription. The TGF-ß/Smad pathway and coregulators Ski and SnoN clearly regulate each other through several positive and negative feedback mechanisms. Thus, these cross-regulatory processes finely modify the TGF-ß signaling outcome as they control the magnitude and duration of the TGF-ß signals. As a result, any alteration in these regulatory mechanisms may lead to disease development. Therefore, the design of targeted therapies to exert tight control of the levels of negative modulators of the TGF-ß pathway, such as Ski and SnoN, is critical to restore cell homeostasis under the specific pathological conditions in which these cofactors are deregulated, such as fibrosis and cancer.

19.
Micromachines (Basel) ; 9(4)2018 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-30424120

RESUMEN

The development of organ-on-chip and biological scaffolds is currently requiring simpler methods for microstructure biocompatible materials in three dimensions, to fabricate structural and functional elements in biomaterials, or modify the physicochemical properties of desired substrates. Aiming at addressing this need, a low-power CD-DVD-Blu-ray laser pickup head was mounted on a programmable three-axis micro-displacement system in order to modify the surface of polymeric materials in a local fashion. Thanks to a specially-designed method using a strongly absorbing additive coating the materials of interest, it has been possible to establish and precisely control processes useful in microtechnology for biomedical applications. The system was upgraded with Blu-ray laser for additive manufacturing and ablation on a single platform. In this work, we present the application of these fabrication techniques to the development of biomimetic cellular culture platforms thanks to the simple integration of several features typically achieved with traditional, less cost-effective microtechnology methods in one step or through replica-molding. Our straightforward approach indeed enables great control of local laser microablation or polymerization for true on-demand biomimetic micropatterned designs in transparent polymers and hydrogels and is allowing integration of microfluidics, microelectronics, surface microstructuring, and transfer of superficial protein micropatterns on a variety of biocompatible materials.

20.
J Tissue Eng ; 8: 2041731417741505, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29225769

RESUMEN

We present the rapid-prototyping of type I collagen micropatterns on poly-dimethylsiloxane substrates for the biomimetic confinement of cells using the combination of a surface oxidation treatment and 3-aminopropyl triethoxysilane silanisation followed by glutaraldehyde crosslinking. The aim of surface treatment is to stabilise microcontact printing transfer of this natural extracellular matrix protein that usually wears out easily from poly-dimethylsiloxane, which is not suitable for biomimetic cell culture platforms and lab-on-chip applications. A low-cost CD-DVD laser was used to etch biomimetic micropatterns into acrylic sheets that were in turn replicated to poly-dimethylsiloxane slabs with the desired features. These stamps were finally inked with type I collagen for microcontact printing transfer on the culture substrates in a simple manner. Human hepatoma cells (HepG2) and rat primary hepatocytes, which do not adhere to bare poly-dimethylsiloxane, were successfully seeded and showed optimal adhesion and survival on simple protein micropatterns with a hepatic cord geometry in order to validate our technique. HepG2 cells also proliferated on the stamps. Soft and stiff poly-dimethylsiloxane layers were also tested to demonstrate that our cost-effective process is compatible with biomimetic organ-on-chip technology integrating tunable stiffness with a potential application to drug testing probes development where such cells are commonly used.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda