Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Microbiome ; 10(1): 237, 2022 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-36566218

RESUMO

BACKGROUND: The Arctic Ocean receives massive freshwater input and a correspondingly large amount of humic-rich organic matter of terrestrial origin. Global warming, permafrost melt, and a changing hydrological cycle will contribute to an intensification of terrestrial organic matter release to the Arctic Ocean. Although considered recalcitrant to degradation due to complex aromatic structures, humic substances can serve as substrate for microbial growth in terrestrial environments. However, the capacity of marine microbiomes to process aromatic-rich humic substances, and how this processing may contribute to carbon and nutrient cycling in a changing Arctic Ocean, is relatively unexplored. Here, we used a combination of metagenomics and metatranscriptomics to assess the prevalence and diversity of metabolic pathways and bacterial taxa involved in aromatic compound degradation in the salinity-stratified summer waters of the Canada Basin in the western Arctic Ocean. RESULTS: Community-scale meta-omics profiling revealed that 22 complete pathways for processing aromatic compounds were present and expressed in the Canada Basin, including those for aromatic ring fission and upstream funneling pathways to access diverse aromatic compounds of terrestrial origin. A phylogenetically diverse set of functional marker genes and transcripts were associated with fluorescent dissolved organic matter, a component of which is of terrestrial origin. Pathways were common throughout global ocean microbiomes but were more abundant in the Canada Basin. Genome-resolved analyses identified 12 clades of Alphaproteobacteria, including Rhodospirillales, as central contributors to aromatic compound processing. These genomes were mostly restricted in their biogeographical distribution to the Arctic Ocean and were enriched in aromatic compound processing genes compared to their closest relatives from other oceans. CONCLUSION: Overall, the detection of a phylogenetically diverse set of genes and transcripts implicated in aromatic compound processing supports the view that Arctic Ocean microbiomes have the capacity to metabolize humic substances of terrestrial origin. In addition, the demonstration that bacterial genomes replete with aromatic compound degradation genes exhibit a limited distribution outside of the Arctic Ocean suggests that processing humic substances is an adaptive trait of the Arctic Ocean microbiome. Future increases in terrestrial organic matter input to the Arctic Ocean may increase the prominence of aromatic compound processing bacteria and their contribution to Arctic carbon and nutrient cycles. Video Abstract.


Assuntos
Substâncias Húmicas , Microbiota , Substâncias Húmicas/análise , Oceanos e Mares , Bactérias/genética , Carbono , Microbiota/genética
2.
Adv Biosyst ; 4(9): e1900230, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32744807

RESUMO

The functional state of the neurovascular unit (NVU), composed of the blood-brain barrier and the perivasculature that forms a dynamic interface between the blood and the central nervous system (CNS), plays a central role in the control of brain homeostasis and is strongly affected by CNS drugs. Human primary brain microvascular endothelium, astrocyte, pericyte, and neural cell cultures are often used to study NVU barrier functions as well as drug transport and efficacy; however, the proteomic and metabolomic responses of these different cell types are not well characterized. Culturing each cell type separately, using deep coverage proteomic analysis and characterization of the secreted metabolome, as well as measurements of mitochondrial activity, the responses of these cells under baseline conditions and when exposed to the NVU-impairing stimulant methamphetamine (Meth) are analyzed. These studies define the previously unknown metabolic and proteomic profiles of human brain pericytes and lead to improved characterization of the phenotype of each of the NVU cell types as well as cell-specific metabolic and proteomic responses to Meth.


Assuntos
Metaboloma/efeitos dos fármacos , Metanfetamina/farmacologia , Neurônios , Pericitos , Proteoma/efeitos dos fármacos , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Encéfalo/irrigação sanguínea , Encéfalo/citologia , Células Cultivadas , Estimulantes do Sistema Nervoso Central/farmacologia , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Humanos , Metabolômica , Neurônios/citologia , Neurônios/efeitos dos fármacos , Pericitos/citologia , Pericitos/efeitos dos fármacos , Proteoma/análise , Proteômica
3.
Integr Biol (Camb) ; 12(3): 64-79, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32195539

RESUMO

The blood-brain barrier plays a critical role in delivering oxygen and nutrients to the brain while preventing the transport of neurotoxins. Predicting the ability of potential therapeutics and neurotoxicants to modulate brain barrier function remains a challenge due to limited spatial resolution and geometric constraints offered by existing in vitro models. Using soft lithography to control the shape of microvascular tissues, we predicted blood-brain barrier permeability states based on structural changes in human brain endothelial cells. We quantified morphological differences in nuclear, junction, and cytoskeletal proteins that influence, or indicate, barrier permeability. We established a correlation between brain endothelial cell pair structure and permeability by treating cell pairs and tissues with known cytoskeleton-modulating agents, including a Rho activator, a Rho inhibitor, and a cyclic adenosine monophosphate analog. Using this approach, we found that high-permeability cell pairs showed nuclear elongation, loss of junction proteins, and increased actin stress fiber formation, which were indicative of increased contractility. We measured traction forces generated by high- and low-permeability pairs, finding that higher stress at the intercellular junction contributes to barrier leakiness. We further tested the applicability of this platform to predict modulations in brain endothelial permeability by exposing cell pairs to engineered nanomaterials, including gold, silver-silica, and cerium oxide nanoparticles, thereby uncovering new insights into the mechanism of nanoparticle-mediated barrier disruption. Overall, we confirm the utility of this platform to assess the multiscale impact of pharmacological agents or environmental toxicants on blood-brain barrier integrity.


Assuntos
Barreira Hematoencefálica , Encéfalo/irrigação sanguínea , Circulação Cerebrovascular , Microcirculação , Actinas/química , Transporte Biológico , Permeabilidade Capilar , Núcleo Celular/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Citoesqueleto/metabolismo , Dimetilpolisiloxanos , Células Endoteliais/metabolismo , Humanos , Junções Intercelulares/metabolismo , Nanopartículas , Permeabilidade
4.
Nat Biomed Eng ; 4(4): 407-420, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31988458

RESUMO

Organ chips can recapitulate organ-level (patho)physiology, yet pharmacokinetic and pharmacodynamic analyses require multi-organ systems linked by vascular perfusion. Here, we describe an 'interrogator' that employs liquid-handling robotics, custom software and an integrated mobile microscope for the automated culture, perfusion, medium addition, fluidic linking, sample collection and in situ microscopy imaging of up to ten organ chips inside a standard tissue-culture incubator. The robotic interrogator maintained the viability and organ-specific functions of eight vascularized, two-channel organ chips (intestine, liver, kidney, heart, lung, skin, blood-brain barrier and brain) for 3 weeks in culture when intermittently fluidically coupled via a common blood substitute through their reservoirs of medium and endothelium-lined vascular channels. We used the robotic interrogator and a physiological multicompartmental reduced-order model of the experimental system to quantitatively predict the distribution of an inulin tracer perfused through the multi-organ human-body-on-chips. The automated culture system enables the imaging of cells in the organ chips and the repeated sampling of both the vascular and interstitial compartments without compromising fluidic coupling.


Assuntos
Técnicas de Cultura de Células/métodos , Dispositivos Lab-On-A-Chip , Microfluídica/métodos , Robótica/métodos , Barreira Hematoencefálica , Encéfalo , Calibragem , Técnicas de Cultura de Células/instrumentação , Desenho de Equipamento , Coração , Humanos , Intestinos , Rim , Fígado , Pulmão , Robótica/instrumentação , Pele
5.
Nanoscale ; 11(38): 17878-17893, 2019 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-31553035

RESUMO

Engineered nanomaterials (ENMs) are increasingly used in consumer products due to their unique physicochemical properties, but the specific hazards they pose to the structural and functional integrity of endothelial barriers remain elusive. When assessing the effects of ENMs on vascular barrier function, endothelial cell monolayers are commonly used as in vitro models. Monolayer models, however, do not offer a granular understanding of how the structure-function relationships between endothelial cells and tissues are disrupted due to ENM exposure. To address this issue, we developed a micropatterned endothelial cell pair model to quantitatively evaluate the effects of 10 ENMs (8 metal/metal oxides and 2 organic ENMs) on multiple cellular parameters and determine how these parameters correlate to changes in vascular barrier function. This minimalistic approach showed concerted changes in endothelial cell morphology, intercellular junction formation, and cytoskeletal organization due to ENM exposure, which were then quantified and compared to unexposed pairs using a "similarity scoring" method. Using the cell pair model, this study revealed dose-dependent changes in actin organization and adherens junction formation following exposure to representative ENMs (Ag, TiO2 and cellulose nanocrystals), which exhibited trends that correlate with changes in tissue permeability measured using an endothelial monolayer assay. Together, these results demonstrate that we can quantitatively evaluate changes in endothelial architecture emergent from nucleo-cytoskeletal network remodeling using micropatterned cell pairs. The endothelial pair model therefore presents potential applicability as a standardized assay for systematically screening ENMs and other test agents for their cellular-level structural effects on vascular barriers.


Assuntos
Núcleo Celular/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Modelos Biológicos , Nanopartículas/química , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos
6.
Nat Biotechnol ; 36(9): 865-874, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30125269

RESUMO

The neurovascular unit (NVU) regulates metabolic homeostasis as well as drug pharmacokinetics and pharmacodynamics in the central nervous system. Metabolic fluxes and conversions over the NVU rely on interactions between brain microvascular endothelium, perivascular pericytes, astrocytes and neurons, making it difficult to identify the contributions of each cell type. Here we model the human NVU using microfluidic organ chips, allowing analysis of the roles of individual cell types in NVU functions. Three coupled chips model influx across the blood-brain barrier (BBB), the brain parenchymal compartment and efflux across the BBB. We used this linked system to mimic the effect of intravascular administration of the psychoactive drug methamphetamine and to identify previously unknown metabolic coupling between the BBB and neurons. Thus, the NVU system offers an in vitro approach for probing transport, efficacy, mechanism of action and toxicity of neuroactive drugs.


Assuntos
Células Endoteliais/metabolismo , Dispositivos Lab-On-A-Chip , Neurônios/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Humanos , Metanfetamina/farmacologia , Fenótipo
7.
Adv Healthc Mater ; 7(9): e1701175, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29359866

RESUMO

Historically, soy protein and extracts have been used extensively in foods due to their high protein and mineral content. More recently, soy protein has received attention for a variety of its potential health benefits, including enhanced skin regeneration. It has been reported that soy protein possesses bioactive molecules similar to extracellular matrix (ECM) proteins and estrogen. In wound healing, oral and topical soy has been heralded as a safe and cost-effective alternative to animal protein and endogenous estrogen. However, engineering soy protein-based fibrous dressings, while recapitulating ECM microenvironment and maintaining a moist environment, remains a challenge. Here, the development of an entirely plant-based nanofibrous dressing comprised of cellulose acetate (CA) and soy protein hydrolysate (SPH) using rotary jet spinning is described. The spun nanofibers successfully mimic physicochemical properties of the native skin ECM and exhibit a high water retaining capability. In vitro, CA/SPH nanofibers promote fibroblast proliferation, migration, infiltration, and integrin ß1 expression. In vivo, CA/SPH scaffolds accelerate re-epithelialization and epidermal thinning as well as reduce scar formation and collagen anisotropy in a similar fashion to other fibrous scaffolds, but without the use of animal proteins or synthetic polymers. These results affirm the potential of CA/SPH nanofibers as a novel wound dressing.


Assuntos
Bandagens , Materiais Biomiméticos/química , Celulose/química , Matriz Extracelular/química , Nanofibras/química , Pele , Proteínas de Soja/química , Alicerces Teciduais/química , Cicatrização , Ferimentos e Lesões/terapia , Animais , Linhagem Celular , Humanos , Masculino , Camundongos , Ferimentos e Lesões/metabolismo , Ferimentos e Lesões/patologia
8.
Cell Adh Migr ; 11(1): 98-109, 2017 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-27111836

RESUMO

The mechanical properties of living cells reflect their propensity to migrate and respond to external forces. Both cellular and nuclear stiffnesses are strongly influenced by the rigidity of the extracellular matrix (ECM) through reorganization of the cyto- and nucleoskeletal protein connections. Changes in this architectural continuum affect cell mechanics and underlie many pathological conditions. In this context, an accurate and combined quantification of the mechanical properties of both cells and nuclei can contribute to a better understanding of cellular (dys-)function. To address this challenge, we have established a robust method for probing cellular and nuclear deformation during spreading and detachment from micropatterned substrates. We show that (de-)adhesion kinetics of endothelial cells are modulated by substrate stiffness and rely on the actomyosin network. We combined this approach with measurements of cell stiffness by magnetic tweezers to show that relaxation dynamics can be considered as a reliable parameter of cellular pre-stress in adherent cells. During the adhesion stage, large cellular and nuclear deformations occur over a long time span (>60 min). Conversely, nuclear deformation and condensed chromatin are relaxed in a few seconds after detachment. Finally, our results show that accumulation of farnesylated prelamin leads to modifications of the nuclear viscoelastic properties, as reflected by increased nuclear relaxation times. Our method offers an original and non-intrusive way of simultaneously gauging cellular and nuclear mechanics, which can be extended to high-throughput screens of pathological conditions and potential countermeasures.


Assuntos
Núcleo Celular/metabolismo , Citoesqueleto/metabolismo , Células Endoteliais da Veia Umbilical Humana/citologia , Microtecnologia/métodos , Estresse Mecânico , Actomiosina/metabolismo , Fenômenos Biomecânicos , Adesão Celular , Movimento Celular , Forma do Núcleo Celular , Forma Celular , Matriz Extracelular/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Cinética , Lamina Tipo A/metabolismo , Fatores de Tempo
10.
Biomaterials ; 89: 14-24, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26946402

RESUMO

The ability to construct easily in vitro networks of primary neurons organized with imposed topologies is required for neural tissue engineering as well as for the development of neuronal interfaces with desirable characteristics. However, accumulating evidence suggests that the mechanical properties of the culture matrix can modulate important neuronal functions such as growth, extension, branching and activity. Here we designed robust and reproducible laminin-polylysine grid micropatterns on cell culture substrates that have similar biochemical properties but a 100-fold difference in Young's modulus to investigate the role of the matrix rigidity on the formation and activity of cortical neuronal networks. We found that cell bodies of primary cortical neurons gradually accumulate in circular islands, whereas axonal extensions spread on linear tracks to connect circular islands. Our findings indicate that migration of cortical neurons is enhanced on soft substrates, leading to a faster formation of neuronal networks. Furthermore, the pre-synaptic density was two times higher on stiff substrates and consistently the number of action potentials and miniature synaptic currents was enhanced on stiff substrates. Taken together, our results provide compelling evidence to indicate that matrix stiffness is a key parameter to modulate the growth dynamics, synaptic density and electrophysiological activity of cortical neuronal networks, thus providing useful information on scaffold design for neural tissue engineering.


Assuntos
Materiais Biocompatíveis/química , Córtex Cerebelar/citologia , Laminina/química , Rede Nervosa/citologia , Neurônios/citologia , Polilisina/química , Potenciais de Ação , Animais , Adesão Celular , Técnicas de Cultura de Células , Movimento Celular , Células Cultivadas , Módulo de Elasticidade , Ratos , Engenharia Tecidual
11.
J Comp Neurol ; 524(7): 1309-36, 2016 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-26780384

RESUMO

In the brain, extracellular matrix (ECM) components form networks that contribute to structural and functional diversity. Maladaptive remodeling of ECM networks has been reported in neurodegenerative and psychiatric disorders, suggesting that the brain microenvironment is a dynamic structure. A lack of quantitative information about ECM distribution in the brain hinders an understanding of region-specific ECM functions and the role of ECM in health and disease. We hypothesized that each ECM protein as well as specific ECM structures, such as perineuronal nets (PNNs) and interstitial matrix, are differentially distributed throughout the brain, contributing to the unique structure and function in the various regions of the brain. To test our hypothesis, we quantitatively analyzed the distribution, colocalization, and protein expression of aggrecan, brevican, and tenascin-R throughout the rat brain utilizing immunohistochemistry and mass spectrometry analysis and assessed the effect of aggrecan, brevican, and/or tenascin-R on neurite outgrowth in vitro. We focused on aggrecan, brevican, and tenascin-R as they are especially expressed in the mature brain, and have established roles in brain development, plasticity, and neurite outgrowth. The results revealed a differentiated distribution of all three proteins throughout the brain and indicated that their presence significantly reduces neurite outgrowth in a 3D in vitro environment. These results underline the importance of a unique and complex ECM distribution for brain physiology and suggest that encoding the distribution of distinct ECM proteins throughout the brain will aid in understanding their function in physiology and in turn assist in identifying their role in disease. J. Comp. Neurol. 524:1309-1336, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Encéfalo/anatomia & histologia , Encéfalo/metabolismo , Proteínas da Matriz Extracelular/genética , Regulação da Expressão Gênica , Agrecanas/metabolismo , Análise de Variância , Animais , Animais Recém-Nascidos , Brevicam/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Proteínas da Matriz Extracelular/metabolismo , Feminino , Imageamento Tridimensional , Espectrometria de Massas , Rede Nervosa/metabolismo , Neuritos/metabolismo , Neuroimagem , Neurônios/metabolismo , Neurônios/ultraestrutura , Ratos , Ratos Sprague-Dawley , Tubulina (Proteína)/metabolismo
12.
Sci Rep ; 5: 9475, 2015 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-25820512

RESUMO

Although pathological changes in axonal morphology have emerged as important features of traumatic brain injury (TBI), the mechanical vulnerability of the axonal microcompartment relative to the cell body is not well understood. We hypothesized that soma and neurite microcompartments exhibit distinct mechanical behaviors, rendering axons more sensitive to a mechanical injury. In order to test this assumption, we combined protein micropatterns with magnetic tweezer rheology to probe the viscoelastic properties of neuronal microcompartments. Creep experiments revealed two opposite rheological behaviors within cortical neurons: the cell body was soft and characterized by a solid-like response, whereas the neurite compartment was stiffer and viscous-like. By using pharmacological agents, we demonstrated that the nucleus is responsible for the solid-like behavior and the stress-stiffening response of the soma, whereas neurofilaments have a predominant contribution in the viscous behavior of the neurite. Furthermore, we found that the neurite is a mechanosensitive compartment that becomes softer and adopts a pronounced viscous state on soft matrices. Together, these findings highlight the importance of the regionalization of mechanical and rigidity-sensing properties within neuron microcompartments in the preferential damage of axons during traumatic brain injury and into potential mechanisms of axonal outgrowth after injury.


Assuntos
Axônios/metabolismo , Lesões Encefálicas/patologia , Neurônios/metabolismo , Animais , Axônios/patologia , Lesões Encefálicas/metabolismo , Córtex Cerebral/metabolismo , Córtex Cerebral/fisiopatologia , Neuritos/metabolismo , Neuritos/patologia , Neurônios/patologia , Ratos , Reologia
13.
Sci Rep ; 4: 7362, 2014 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-25482017

RESUMO

Increasing evidences show that the actin cytoskeleton is a key parameter of the nuclear remodeling process in response to the modifications of cellular morphology. However, detailed information on the interaction between the actin cytoskeleton and the nuclear lamina was still lacking. We addressed this question by constraining endothelial cells on rectangular fibronectin-coated micropatterns and then using Structured Illumination Microscopy (SIM) to observe the interactions between actin stress fibers, nuclear lamina and LINC complexes at a super-resolution scale. Our results show that tension in apical actin stress fibers leads to deep nuclear indentations that significantly deform the nuclear lamina. Interestingly, indented nuclear zones are characterized by a local enrichment of LINC complexes, which anchor apical actin fibers to the nuclear lamina. Moreover, our findings indicate that nuclear indentations induce the formation of segregated domains of condensed chromatin. However, nuclear indentations and condensed chromatin domains are not irreversible processes and both can relax in absence of tension in apical actin stress fibers.


Assuntos
Citoesqueleto/metabolismo , Microscopia/métodos , Lâmina Nuclear/metabolismo , Núcleo Celular , Cromatina/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Ligação Proteica , Transporte Proteico , Fibras de Estresse/metabolismo
14.
J Vis Exp ; (90)2014 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-25225964

RESUMO

It is now well established that many cellular functions are regulated by interactions of cells with physicochemical and mechanical cues of their extracellular matrix (ECM) environment. Eukaryotic cells constantly sense their local microenvironment through surface mechanosensors to transduce physical changes of ECM into biochemical signals, and integrate these signals to achieve specific changes in gene expression. Interestingly, physicochemical and mechanical parameters of the ECM can couple with each other to regulate cell fate. Therefore, a key to understanding mechanotransduction is to decouple the relative contribution of ECM cues on cellular functions. Here we present a detailed experimental protocol to rapidly and easily generate biologically relevant hydrogels for the independent tuning of mechanotransduction cues in vitro. We chemically modified polyacrylamide hydrogels (PAAm) to surmount their intrinsically non-adhesive properties by incorporating hydroxyl-functionalized acrylamide monomers during the polymerization. We obtained a novel PAAm hydrogel, called hydroxy-PAAm, which permits immobilization of any desired nature of ECM proteins. The combination of hydroxy-PAAm hydrogels with microcontact printing allows to independently control the morphology of single-cells, the matrix stiffness, the nature and the density of ECM proteins. We provide a simple and rapid method that can be set up in every biology lab to study in vitro cell mechanotransduction processes. We validate this novel two-dimensional platform by conducting experiments on endothelial cells that demonstrate a mechanical coupling between ECM stiffness and the nucleus.


Assuntos
Resinas Acrílicas/química , Células Endoteliais/citologia , Hidrogéis/química , Resinas Acrílicas/síntese química , Núcleo Celular/química , Núcleo Celular/metabolismo , Células Endoteliais/química , Células Endoteliais/metabolismo , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/química , Proteínas da Matriz Extracelular/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Hidrogéis/síntese química , Proteínas Imobilizadas/química , Mecanotransdução Celular/fisiologia , Impressão/métodos , Análise de Célula Única/métodos
15.
Methods Cell Biol ; 121: 33-48, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24560501

RESUMO

This protocol describes a simple method to deposit protein micropatterns over a wide range of culture substrate stiffness (three orders of magnitude) by using two complementary polymeric substrates. In the first part, we introduce a novel polyacrylamide hydrogel, called hydroxy-polyacrylamide (PAAm), that permits to surmount the intrinsically nonadhesive properties of polyacrylamide with minimal requirements in cost or expertize. We present a protocol for tuning easily the rigidity of "soft" hydroxy-PAAm hydrogels between ~0.5 and 50 kPa and a micropatterning method to locally deposit protein micropatterns on these hydrogels. In a second part, we describe a protocol for tuning the rigidity of "stiff" silicone elastomers between ~100 and 1000 kPa and printing efficiently proteins from the extracellular matrix. Finally, we investigate the effect of the matrix rigidity on the nucleus of primary endothelial cells by tuning the rigidity of both polymeric substrates. We envision that the complementarity of these two polymeric substrates, combined with an efficient microprinting technique, can be further developed in the future as a powerful mechanobiology platform to investigate in vitro the effect of mechanotransduction cues on cellular functions, gene expression, and stem cell differentiation.


Assuntos
Resinas Acrílicas/química , Materiais Revestidos Biocompatíveis , Módulo de Elasticidade/fisiologia , Hidrogéis/química , Elastômeros de Silicone/química , Células 3T3 , Animais , Adesão Celular/fisiologia , Técnicas de Cultura de Células , Linhagem Celular , Microambiente Celular , Matriz Extracelular/fisiologia , Mecanotransdução Celular/fisiologia , Camundongos , Impressão , Proteínas/química , Ratos , Estresse Mecânico , Propriedades de Superfície
16.
Lab Chip ; 13(5): 777-80, 2013 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-23334710

RESUMO

Physico-chemical and biochemical factors in the local cellular microenvironment are known to impact on multiple aspects of cell behaviour through specific signal pathways. These mechanotransduction cues can couple each other to regulate cell fate, and it remains unclear whether mechanotransduction in different contexts shares common mechanisms. Undoubtedly, a challenge will involve the further characterization of such cooperative mechanisms, as well as clearly defining the individual role of each mechanical and biochemical parameter. To control these mechanotransduction cues in an independent manner, we developed a simple and efficient strategy to immobilize any desired nature of proteins on polyacrylamide hydrogels and independently control various parameters of the cell microenvironment, such as matrix stiffness, cell-binding ligand density and confined adhesiveness. This novel platform is validated by conducting single-cell experiments and opens a broad avenue for studying complex interplays involved in mechanotransduction with a facile and versatile approach.


Assuntos
Resinas Acrílicas/química , Hidrogéis/química , Mecanotransdução Celular , Resinas Acrílicas/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Proteínas da Matriz Extracelular/química , Proteínas da Matriz Extracelular/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Hidrogéis/toxicidade
17.
Nat Commun ; 3: 671, 2012 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-22334074

RESUMO

Growing evidence suggests that cytoplasmic actin filaments are essential factors in the modulation of nuclear shape and function. However, the mechanistic understanding of the internal orchestration between cell and nuclear shape is still lacking. Here we show that orientation and deformation of the nucleus are regulated by lateral compressive forces driven by tension in central actomyosin fibres. By using a combination of micro-manipulation tools, our study reveals that tension in central stress fibres is gradually generated by anisotropic force contraction dipoles, which expand as the cell elongates and spreads. Our findings indicate that large-scale cell shape changes induce a drastic condensation of chromatin and dramatically affect cell proliferation. On the basis of these findings, we propose a simple mechanical model that quantitatively accounts for our experimental data and provides a conceptual framework for the mechanistic coordination between cell and nuclear shape.


Assuntos
Núcleo Celular/metabolismo , Células Endoteliais/citologia , Actinas/química , Actomiosina/química , Biofísica/métodos , Proliferação de Células , Forma Celular , Cromatina/química , Força Compressiva , Citoesqueleto , Adesões Focais , Humanos , Imageamento Tridimensional , Micromanipulação , Modelos Biológicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA