Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Elife ; 102021 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-34378534

RESUMO

Traditional drug safety assessment often fails to predict complications in humans, especially when the drug targets the immune system. Here, we show the unprecedented capability of two human Organs-on-Chips to evaluate the safety profile of T-cell bispecific antibodies (TCBs) targeting tumor antigens. Although promising for cancer immunotherapy, TCBs are associated with an on-target, off-tumor risk due to low levels of expression of tumor antigens in healthy tissues. We leveraged in vivo target expression and toxicity data of TCBs targeting folate receptor 1 (FOLR1) or carcinoembryonic antigen (CEA) to design and validate human immunocompetent Organs-on-Chips safety platforms. We discovered that the Lung-Chip and Intestine-Chip could reproduce and predict target-dependent TCB safety liabilities, based on sensitivity to key determinants thereof, such as target expression and antibody affinity. These novel tools broaden the research options available for mechanistic understandings of engineered therapeutic antibodies and assessing safety in tissues susceptible to adverse events.


Assuntos
Anticorpos Biespecíficos/efeitos adversos , Dispositivos Lab-On-A-Chip/estatística & dados numéricos , Linfócitos T/imunologia , Animais , Feminino , Células HEK293 , Células HeLa , Humanos , Imunoterapia/métodos , Camundongos
2.
Nature ; 585(7826): 574-578, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32939089

RESUMO

Epithelial organoids, such as those derived from stem cells of the intestine, have great potential for modelling tissue and disease biology1-4. However, the approaches that are used at present to derive these organoids in three-dimensional matrices5,6 result in stochastically developing tissues with a closed, cystic architecture that restricts lifespan and size, limits experimental manipulation and prohibits homeostasis. Here, by using tissue engineering and the intrinsic self-organization properties of cells, we induce intestinal stem cells to form tube-shaped epithelia with an accessible lumen and a similar spatial arrangement of crypt- and villus-like domains to that in vivo. When connected to an external pumping system, the mini-gut tubes are perfusable; this allows the continuous removal of dead cells to prolong tissue lifespan by several weeks, and also enables the tubes to be colonized with microorganisms for modelling host-microorganism interactions. The mini-intestines include rare, specialized cell types that are seldom found in conventional organoids. They retain key physiological hallmarks of the intestine and have a notable capacity to regenerate. Our concept for extrinsically guiding the self-organization of stem cells into functional organoids-on-a-chip is broadly applicable and will enable the attainment of more physiologically relevant organoid shapes, sizes and functions.


Assuntos
Homeostase , Intestinos/embriologia , Morfogênese , Organoides/embriologia , Alicerces Teciduais , Animais , Padronização Corporal , Diferenciação Celular , Linhagem da Célula , Cryptosporidium parvum/patogenicidade , Células-Tronco Embrionárias Humanas/citologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Intestinos/citologia , Intestinos/parasitologia , Intestinos/patologia , Camundongos , Modelos Biológicos , Organoides/citologia , Organoides/parasitologia , Organoides/patologia , Regeneração , Medicina Regenerativa , Células-Tronco , Técnicas de Cultura de Tecidos/métodos , Engenharia Tecidual
3.
Lab Chip ; 20(18): 3365-3374, 2020 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-32761043

RESUMO

The multiphasic etiology of tissue inflammation and the fundamental immunological differences between species render inflammatory pathologies difficult to recapitulate in animal models, and account for the paucity of therapies that are successfully translated from rodents to humans. Here, we present a human-relevant organ-on-a-chip platform for experimental inflammatory diseases. We created an immunocompetent in vitro gut model by incorporating intestinal epithelial and immune cells into microfluidic chambers that permit cell movement across an extracellular matrix (ECM) and fluidic channels. This is the first model that integrates a mucosal barrier, a three-dimensional ECM, resident and infiltrating immune cells, and simulates a functional crosstalk that ultimately triggers cellular processes representative of inflammation. Under homeostatic conditions, enterocytes form a tight epithelium and subepithelial macrophages are non-activated. Introduction of pro-inflammatory mediators triggers macrophage activation and inflammation-induced intestinal barrier leakiness. Neutrophils in a parallel, matrix-separated non-epithelial channel are attracted by such a pro-inflammatory microenvironment and migrate through the extracellular matrix, further exacerbating tissue inflammation and damage. With this model, we provide the foundations to recapitulate and investigate the onset of tissue inflammation in a controlled, human-relevant system.


Assuntos
Inflamação , Dispositivos Lab-On-A-Chip , Animais , Matriz Extracelular , Homeostase , Macrófagos
4.
Nat Biomed Eng ; 4(9): 863-874, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32514094

RESUMO

Stem-cell-derived epithelial organoids are routinely used for the biological and biomedical modelling of tissues. However, the complexity, lack of standardization and quality control of stem cell culture in solid extracellular matrices hampers the routine use of the organoids at the industrial scale. Here, we report the fabrication of microengineered cell culture devices and scalable and automated methods for suspension culture and real-time analysis of thousands of individual gastrointestinal organoids trapped in microcavity arrays within a polymer-hydrogel substrate. The absence of a solid matrix substantially reduces organoid heterogeneity, which we show for mouse and human gastrointestinal organoids. We use the devices to screen for anticancer drug candidates with patient-derived colorectal cancer organoids, and apply high-content image-based phenotypic analyses to reveal insights into mechanisms of drug action. The scalable organoid-culture technology should facilitate the use of organoids in drug development and diagnostics.


Assuntos
Técnicas de Cultura de Células/métodos , Organoides/citologia , Células-Tronco/citologia , Animais , Agregação Celular , Células Cultivadas , Dimetilpolisiloxanos/química , Avaliação Pré-Clínica de Medicamentos , Ensaios de Triagem em Larga Escala , Humanos , Hidrogéis/química , Intestinos/citologia , Camundongos , Organogênese , Organoides/efeitos dos fármacos , Organoides/crescimento & desenvolvimento
5.
J Clin Invest ; 130(3): 1199-1216, 2020 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-32015230

RESUMO

Mutations in APC promote colorectal cancer (CRC) progression through uncontrolled WNT signaling. Patients with desmoplastic CRC have a significantly worse prognosis and do not benefit from chemotherapy, but the mechanisms underlying the differential responses of APC-mutant CRCs to chemotherapy are not well understood. We report that expression of the transcription factor prospero homeobox 1 (PROX1) was reduced in desmoplastic APC-mutant human CRCs. In genetic Apc-mutant mouse models, loss of Prox1 promoted the growth of desmoplastic, angiogenic, and immunologically silent tumors through derepression of Mmp14. Although chemotherapy inhibited Prox1-proficient tumors, it promoted further stromal activation, angiogenesis, and invasion in Prox1-deficient tumors. Blockade of vascular endothelial growth factor A (VEGFA) and angiopoietin-2 (ANGPT2) combined with CD40 agonistic antibodies promoted antiangiogenic and immunostimulatory reprogramming of Prox1-deficient tumors, destroyed tumor fibrosis, and unleashed T cell-mediated killing of cancer cells. These results pinpoint the mechanistic basis of chemotherapy-induced hyperprogression and illustrate a therapeutic strategy for chemoresistant and desmoplastic CRCs.


Assuntos
Inibidores da Angiogênese/farmacologia , Antineoplásicos Imunológicos/farmacologia , Neoplasias Colorretais , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Imunoterapia , Neovascularização Patológica , Proteína da Polipose Adenomatosa do Colo/genética , Proteína da Polipose Adenomatosa do Colo/imunologia , Angiopoietina-2/genética , Angiopoietina-2/imunologia , Animais , Linhagem Celular , Neoplasias Colorretais/irrigação sanguínea , Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/terapia , Resistencia a Medicamentos Antineoplásicos/genética , Resistencia a Medicamentos Antineoplásicos/imunologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/imunologia , Humanos , Metaloproteinase 14 da Matriz/genética , Metaloproteinase 14 da Matriz/imunologia , Camundongos , Neoplasias Experimentais/irrigação sanguínea , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Neoplasias Experimentais/terapia , Neovascularização Patológica/genética , Neovascularização Patológica/imunologia , Neovascularização Patológica/terapia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/imunologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/imunologia
6.
Nat Protoc ; 12(11): 2263-2274, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28981121

RESUMO

Growing cells within an extracellular matrix-like 3D gel is required for, or can improve, the growth of many cell types ex vivo. Here, we describe a protocol for the generation of well-defined matrices for the culture of intestinal stem cells (ISCs) and intestinal organoids. These matrices comprise a poly(ethylene glycol) (PEG) hydrogel backbone functionalized with minimal adhesion cues including RGD (Arg-Gly-Asp), which is sufficient for ISC expansion, and laminin-111, which is required for organoid formation. As such, the hydrogels present a defined and reproducible, but also tunable, environment, allowing researches to manipulate physical and chemical parameters, and examine their influence on ISC and organoid growth. Hydrogels are formed by an enzymatic cross-linking reaction of multiarm PEG precursors bearing glutamine- and lysine-containing peptides. PEG precursors containing either stable or hydrolytically degradable moieties are used to produce mechanically softening hydrogels, which are used for the expansion of ISCs or the formation of organoids, respectively. We also provide protocols for immunofluorescence analysis of cellular structures grown within these matrices, as well as for their dissociation and retrieval of cells for downstream use. Hydrogel precursors can be produced and their mechanical properties characterized to ascertain stiffness within 5-7 d. Hydrogel formation for ISC expansion or organoid formation takes 1-2 h. The materials described here can be readily adapted for the culture of other types of normal or transformed organoid structures.


Assuntos
Hidrogel de Polietilenoglicol-Dimetacrilato/química , Intestinos/citologia , Organoides , Células-Tronco , Técnicas de Cultura de Tecidos/métodos , Animais , Células Cultivadas , Reagentes de Ligações Cruzadas , Hidrogel de Polietilenoglicol-Dimetacrilato/síntese química , Laminina/metabolismo , Camundongos , Oligopeptídeos/metabolismo , Organoides/citologia , Organoides/fisiologia , Células-Tronco/citologia , Células-Tronco/fisiologia
7.
Stem Cells Int ; 2017: 8387297, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29081810

RESUMO

Intestinal stem cells are located at the base of the crypts and are surrounded by a complex structure called niche. This environment is composed mainly of epithelial cells and stroma which provides signals that govern cell maintenance, proliferation, and differentiation. Understanding how the niche regulates stem cell fate by controlling developmental signaling pathways will help us to define how stem cells choose between self-renewal and differentiation and how they maintain their undifferentiated state. Tractable in vitro assay systems, which reflect the complexity of the in vivo situation but provide higher level of control, would likely be crucial in identifying new players and mechanisms controlling stem cell function. Knowledge of the intestinal stem cell niche gathered from both in vivo and novel in vitro models may help us improve therapies for tumorigenesis and intestinal damage and make autologous intestinal transplants a feasible clinical practice.

8.
Nature ; 539(7630): 560-564, 2016 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-27851739

RESUMO

Epithelial organoids recapitulate multiple aspects of real organs, making them promising models of organ development, function and disease. However, the full potential of organoids in research and therapy has remained unrealized, owing to the poorly defined animal-derived matrices in which they are grown. Here we used modular synthetic hydrogel networks to define the key extracellular matrix (ECM) parameters that govern intestinal stem cell (ISC) expansion and organoid formation, and show that separate stages of the process require different mechanical environments and ECM components. In particular, fibronectin-based adhesion was sufficient for ISC survival and proliferation. High matrix stiffness significantly enhanced ISC expansion through a yes-associated protein 1 (YAP)-dependent mechanism. ISC differentiation and organoid formation, on the other hand, required a soft matrix and laminin-based adhesion. We used these insights to build a fully defined culture system for the expansion of mouse and human ISCs. We also produced mechanically dynamic matrices that were initially optimal for ISC expansion and subsequently permissive to differentiation and intestinal organoid formation, thus creating well-defined alternatives to animal-derived matrices for the culture of mouse and human stem-cell-derived organoids. Our approach overcomes multiple limitations of current organoid cultures and greatly expands their applicability in basic and clinical research. The principles presented here can be extended to identify designer matrices that are optimal for long-term culture of other types of stem cells and organoids.


Assuntos
Técnicas de Cultura de Células/métodos , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Intestinos/citologia , Organoides/citologia , Organoides/crescimento & desenvolvimento , Células-Tronco/citologia , Técnicas de Cultura de Tecidos/métodos , Animais , Adesão Celular , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Forma Celular , Fibronectinas/metabolismo , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato/síntese química , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Camundongos , Proteólise , Nicho de Células-Tronco
9.
Sci Rep ; 5: 11458, 2015 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-26165921

RESUMO

Collective cell migration drives tissue remodeling during development, wound repair, and metastatic invasion. The physical mechanisms by which cells move cohesively through dense three-dimensional (3D) extracellular matrix (ECM) remain incompletely understood. Here, we show directly that migration of multicellular cohorts through collagenous matrices occurs via a dynamic pulling mechanism, the nature of which had only been inferred previously in 3D. Tensile forces increase at the invasive front of cohorts, serving a physical, propelling role as well as a regulatory one by conditioning the cells and matrix for further extension. These forces elicit mechanosensitive signaling within the leading edge and align the ECM, creating microtracks conducive to further migration. Moreover, cell movements are highly correlated and in phase with ECM deformations. Migrating cohorts use spatially localized, long-range forces and consequent matrix alignment to navigate through the ECM. These results suggest biophysical forces are critical for 3D collective migration.


Assuntos
Técnicas de Cultura de Células , Matriz Extracelular/metabolismo , Animais , Movimento Celular , Células Cultivadas , Colágeno Tipo I/química , Géis/química , Humanos , Processamento de Imagem Assistida por Computador , Camundongos , Microscopia Confocal , Modelos Biológicos , Transdução de Sinais , Resistência à Tração , Imagem com Lapso de Tempo , Transativadores/antagonistas & inibidores , Transativadores/genética , Transativadores/metabolismo
10.
Methods Mol Biol ; 1189: 191-206, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25245695

RESUMO

Several biological processes, including cell migration, tissue morphogenesis, and cancer metastasis, are fundamentally physical in nature; each implicitly involves deformations driven by mechanical forces. Traction force microscopy (TFM) was initially developed to quantify the forces exerted by individual isolated cells in two-dimensional (2D) culture. Here, we extend this technique to estimate the traction forces generated by engineered three-dimensional (3D) epithelial tissues embedded within a surrounding extracellular matrix (ECM). This technique provides insight into the physical mechanisms that underlie tissue morphogenesis in 3D.


Assuntos
Epitélio/fisiologia , Imageamento Tridimensional , Microscopia de Força Atômica , Engenharia Tecidual/métodos , Animais , Dimetilpolisiloxanos/química , Fluorescência , Microesferas , Morfogênese , Imagem com Lapso de Tempo
11.
Proc Natl Acad Sci U S A ; 109(48): 19632-7, 2012 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-23150585

RESUMO

Breast tumor development is regulated in part by cues from the local microenvironment, including interactions with neighboring nontumor cells as well as the ECM. Studies using homogeneous populations of breast cancer cell lines cultured in 3D ECM have shown that increased ECM stiffness stimulates tumor cell invasion. However, at early stages of breast cancer development, malignant cells are surrounded by normal epithelial cells, which have been shown to exert a tumor-suppressive effect on cocultured cancer cells. Here we explored how the biophysical characteristics of the host microenvironment affect the proliferative and invasive tumor phenotype of the earliest stages of tumor development, by using a 3D microfabrication-based approach to engineer ducts composed of normal mammary epithelial cells that contained a single tumor cell. We found that the phenotype of the tumor cell was dictated by its position in the duct: proliferation and invasion were enhanced at the ends and blocked when the tumor cell was located elsewhere within the tissue. Regions of invasion correlated with high endogenous mechanical stress, as shown by finite element modeling and bead displacement experiments, and modulating the contractility of the host epithelium controlled the subsequent invasion of tumor cells. Combining microcomputed tomographic analysis with finite element modeling suggested that predicted regions of high mechanical stress correspond to regions of tumor formation in vivo. This work suggests that the mechanical tone of nontumorigenic host epithelium directs the phenotype of tumor cells and provides additional insight into the instructive role of the mechanical tumor microenvironment.


Assuntos
Neoplasias da Mama/patologia , Invasividade Neoplásica , Matriz Extracelular/patologia , Feminino , Adesões Focais , Humanos , Integrinas/metabolismo , Glândulas Mamárias Humanas/patologia
12.
Biophys J ; 103(1): 152-62, 2012 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-22828342

RESUMO

Understanding how physical signals guide biological processes requires qualitative and quantitative knowledge of the mechanical forces generated and sensed by cells in a physiologically realistic three-dimensional (3D) context. Here, we used computational modeling and engineered epithelial tissues of precise geometry to define the experimental parameters that are required to measure directly the mechanical stress profile of 3D tissues embedded within native type I collagen. We found that to calculate the stresses accurately in these settings, we had to account for mechanical heterogeneities within the matrix, which we visualized and quantified using confocal reflectance and atomic force microscopy. Using this technique, we were able to obtain traction forces at the epithelium-matrix interface, and to resolve and quantify patterns of mechanical stress throughout the surrounding matrix. We discovered that whereas single cells generate tension by contracting and pulling on the matrix, the contraction of multicellular tissues can also push against the matrix, causing emergent compression. Furthermore, tissue geometry defines the spatial distribution of mechanical stress across the epithelium, which communicates mechanically over distances spanning hundreds of micrometers. Spatially resolved mechanical maps can provide insight into the types and magnitudes of physical parameters that are sensed and interpreted by multicellular tissues during normal and pathological processes.


Assuntos
Epitélio/fisiologia , Estresse Mecânico , Engenharia Tecidual , Animais , Fenômenos Biomecânicos , Colágeno Tipo I/química , Epitélio/química , Matriz Extracelular/química , Camundongos , Microscopia de Força Atômica
13.
Cancer Microenviron ; 5(1): 29-38, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21748438

RESUMO

Epithelial-mesenchymal transition (EMT) is a phenotypic shift wherein epithelial cells lose or loosen attachments to their neighbors and assume a mesenchymal-like morphology. EMT drives a variety of developmental processes, but may also be adopted by tumor cells during neoplastic progression. EMT is regulated by both biochemical and physical signals from the microenvironment, including mechanical stress, which is increasingly recognized to play a major role in development and disease progression. Biological systems generate, transmit and concentrate mechanical stress into spatial patterns; these gradients in mechanical stress may serve to spatially pattern developmental and pathologic EMTs. Here we review how epithelial tissues generate and respond to mechanical stress gradients, and highlight the mechanisms by which mechanical stress regulates and patterns EMT.

14.
Nat Rev Mol Cell Biol ; 12(9): 581-93, 2011 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-21829222

RESUMO

The mammary gland undergoes a spectacular series of changes as it develops, and maintains a remarkable capacity to remodel and regenerate for several decades. Mammary morphogenesis has been investigated for over 100 years, motivated by the dairy industry and cancer biologists. Over the past decade, the gland has emerged as a major model system in its own right for understanding the cell biology of tissue morphogenesis. Multiple signalling pathways from several cell types are orchestrated together with mechanical cues and cell rearrangements to establish the pattern of the mammary gland. The integrated mechanical and molecular pathways that control mammary morphogenesis have implications for the developmental regulation of other epithelial organs.


Assuntos
Glândulas Mamárias Humanas/anatomia & histologia , Glândulas Mamárias Humanas/fisiologia , Morfogênese/fisiologia , Animais , Feminino , Humanos , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/fisiologia , Glândulas Mamárias Humanas/metabolismo , Modelos Biológicos , Morfogênese/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Integração de Sistemas
15.
EMBO J ; 30(13): 2662-74, 2011 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-21610693

RESUMO

Several E-box-binding transcription factors regulate individual and collective cell migration and enhance the motility of epithelial cells by promoting epithelial-mesenchymal transition (EMT). Here, we characterized the role of a subset of these transcription factors and the EMT proteome in branching morphogenesis of mammary epithelial tissues using a three-dimensional organotypic culture model of the mammary duct. We found that the transcription factors Snail1, Snail2, and E47 were transiently upregulated at branch sites; decreasing the expression of these transcription factors inhibited branching. Conversely, ectopic expression of Snail1, Snail2, and E47 induced branching in the absence of exogenous stimuli. These changes correlated with the expression of mesenchymal markers and repression of E-cadherin, which was essential for branching. Snail1 and Snail2 also promoted cell survival at branch sites, but this was not sufficient to induce branching. These findings indicate that Snail1, Snail2, and E47 can promote collective migration during branching morphogenesis of mammary epithelial tissues through key regulators of EMT.


Assuntos
Glândulas Mamárias Animais/crescimento & desenvolvimento , Morfogênese/genética , Fator 3 de Transcrição/fisiologia , Fatores de Transcrição/fisiologia , Animais , Movimento Celular/genética , Movimento Celular/fisiologia , Células Cultivadas , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transição Epitelial-Mesenquimal/genética , Transição Epitelial-Mesenquimal/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Técnicas de Silenciamento de Genes , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/metabolismo , Camundongos , Modelos Biológicos , Morfogênese/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , Fatores de Transcrição da Família Snail , Fator 3 de Transcrição/antagonistas & inibidores , Fator 3 de Transcrição/genética , Fator 3 de Transcrição/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
16.
Artigo em Inglês | MEDLINE | ID: mdl-20890968

RESUMO

Invertebrates and vertebrates use branching morphogenesis to build epithelial trees to maximize the surface area of organs within a given volume. Several molecular regulators of branching have recently been discovered, a number of which are conserved across different organs and species. Signals that control branching at the cellular and tissue levels are also starting to emerge, and are rapidly unveiling the physical nature of branch development. Here we discuss the molecular, cellular, and physical processes that govern branch formation, and highlight the major outstanding questions in the field.


Assuntos
Morfogênese , Organogênese , Animais , Drosophila/embriologia , Drosophila/crescimento & desenvolvimento , Epitélio/fisiologia , Fatores de Crescimento de Fibroblastos/metabolismo , Fatores de Crescimento de Fibroblastos/fisiologia , Transdução de Sinais
17.
Birth Defects Res C Embryo Today ; 90(3): 193-202, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20860059

RESUMO

Embryonic development is a physical process during which groups of cells are sculpted into functional organs. The mechanical properties of tissues and the forces exerted on them serve as epigenetic regulators of morphogenesis. Understanding these mechanobiological effects in the embryo requires new experimental approaches. Here we focus on branching of the lung airways and bending of the heart tube to describe examples of mechanical and physical cues that guide cell fate decisions and organogenesis. We highlight recent technological advances to measure tissue elasticity and endogenous mechanical stresses in real time during organ development. We also discuss recent progress in manipulating forces in intact embryos.


Assuntos
Citoesqueleto/genética , Desenvolvimento Embrionário , Morfogênese , Organogênese , Fenômenos Biomecânicos , Diferenciação Celular , Epigenômica , Coração/embriologia , Humanos , Pulmão/embriologia , Microscopia de Força Atômica , Modelos Biológicos
18.
Integr Biol (Camb) ; 2(9): 424-34, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20717570

RESUMO

Spatial patterning of cell behaviors establishes the regional differences within tissues that collectively develop branched organs into their characteristic treelike shapes. Here we show that the pattern of branching morphogenesis of three-dimensional (3D) engineered epithelial tissues is controlled in part by gradients of endogenous mechanical stress. We used microfabrication to build model mammary epithelial tissues of defined geometry that branched in a stereotyped pattern when induced with growth factors. Branches initiated from sites of high mechanical stress within the tissues, as predicted numerically and measured directly using 3D traction force microscopy. Branch sites were defined by activation of focal adhesion kinase (FAK), inhibition of which disrupted morphogenesis. Stress, FAK activation, and branching were all altered by manipulating cellular contractility, matrix stiffness, intercellular cohesion and tissue geometry. These data suggest that the pattern and magnitude of mechanical stress across epithelial tissues cooperate with biochemical signals to specify branching pattern.


Assuntos
Modelos Biológicos , Morfogênese/fisiologia , Animais , Fenômenos Biomecânicos , Padronização Corporal/fisiologia , Linhagem Celular , Epitélio/crescimento & desenvolvimento , Epitélio/fisiologia , Matriz Extracelular/fisiologia , Feminino , Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/fisiologia , Camundongos , Estresse Mecânico , Imagem com Lapso de Tempo , Engenharia Tecidual
19.
J Cell Biochem ; 110(1): 44-51, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20336666

RESUMO

Epithelial-mesenchymal transition (EMT) is a phenotypic change in which epithelial cells detach from their neighbors and become motile. Whereas soluble signals such as growth factors and cytokines are responsible for stimulating EMT, here we show that gradients of mechanical stress define the spatial locations at which EMT occurs. When treated with transforming growth factor (TGF)-beta, cells at the corners and edges of square mammary epithelial sheets expressed EMT markers, whereas those in the center did not. Changing the shape of the epithelial sheet altered the spatial pattern of EMT. Traction force microscopy and finite element modeling demonstrated that EMT-permissive regions experienced the highest mechanical stress. Myocardin-related transcription factor (MRTF)-A was localized to the nuclei of cells located in high-stress regions, and inhibiting cytoskeletal tension or MRTF-A expression abrogated the spatial patterning of EMT. These data suggest a causal role for tissue geometry and endogenous mechanical stresses in the spatial patterning of EMT.


Assuntos
Células Epiteliais/citologia , Células Epiteliais/metabolismo , Espaço Extracelular/metabolismo , Mecanotransdução Celular , Mesoderma/citologia , Mesoderma/metabolismo , Animais , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Células Epiteliais/efeitos dos fármacos , Espaço Extracelular/efeitos dos fármacos , Humanos , Contração Isométrica/efeitos dos fármacos , Mecanotransdução Celular/efeitos dos fármacos , Mesoderma/efeitos dos fármacos , Camundongos , Transporte Proteico/efeitos dos fármacos , Estresse Mecânico , Transativadores/metabolismo , Fator de Crescimento Transformador beta/farmacologia
20.
Cytokine Growth Factor Rev ; 20(5-6): 459-65, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19896886

RESUMO

Normal tissue development and function are regulated by the interplay between cells and their surrounding extracellular matrix (ECM). The ECM provides biochemical and mechanical contextual information that is conveyed from the cell membrane through the cytoskeleton to the nucleus to direct cell phenotype. Cells, in turn, remodel the ECM and thereby sculpt their local microenvironment. Here we review the mechanisms by which cells interact with, respond to, and influence the ECM, with particular emphasis placed on the role of this bidirectional communication during tissue morphogenesis. We also discuss the implications for successful engineering of functional tissues ex vivo.


Assuntos
Matriz Extracelular/fisiologia , Morfogênese/fisiologia , Transdução de Sinais/fisiologia , Animais , Diferenciação Celular/fisiologia , Proliferação de Células , Matriz Extracelular/metabolismo , Humanos , Mecanotransdução Celular/fisiologia , Modelos Biológicos , Engenharia Tecidual/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA