Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Proc Natl Acad Sci U S A ; 116(9): 3502-3507, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30755531

ABSTRACT

Breast cancer development is associated with increasing tissue stiffness over years. To more accurately mimic the onset of gradual matrix stiffening, which is not feasible with conventional static hydrogels, mammary epithelial cells (MECs) were cultured on methacrylated hyaluronic acid hydrogels whose stiffness can be dynamically modulated from "normal" (<150 Pascals) to "malignant" (>3,000 Pascals) via two-stage polymerization. MECs form and remain as spheroids, but begin to lose epithelial characteristics and gain mesenchymal morphology upon matrix stiffening. However, both the degree of matrix stiffening and culture time before stiffening play important roles in regulating this conversion as, in both cases, a subset of mammary spheroids remained insensitive to local matrix stiffness. This conversion depended neither on colony size nor cell density, and MECs did not exhibit "memory" of prior niche when serially cultured through cycles of compliant and stiff matrices. Instead, the transcription factor Twist1, transforming growth factor ß (TGFß), and YAP activation appeared to modulate stiffness-mediated signaling; when stiffness-mediated signals were blocked, collective MEC phenotypes were reduced in favor of single MECs migrating away from spheroids. These data indicate a more complex interplay of time-dependent stiffness signaling, spheroid structure, and soluble cues that regulates MEC plasticity than suggested by previous models.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Breast Neoplasms/genetics , Mechanotransduction, Cellular/genetics , Phosphoproteins/genetics , Transforming Growth Factor beta/genetics , Twist-Related Protein 1/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Hydrogels/chemistry , Paracrine Communication/genetics , Signal Transduction/genetics , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Transcription Factors , YAP-Signaling Proteins
2.
J Biomech Eng ; 138(2): 021003, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26746491

ABSTRACT

Hyaluronic acid (HA) is a commonly used natural polymer for cell scaffolding. Modification by methacrylate allows it to be polymerized by free radicals via addition of an initiator, e.g., light-sensitive Irgacure, to form a methacrylated hyaluronic acid (MeHA) hydrogel. Light-activated crosslinking can be used to control the degree of polymerization, and sequential polymerization steps allow cells plated onto or in the hydrogel to initially feel a soft and then a stiff matrix. Here, the elastic modulus of MeHA hydrogels was systematically analyzed by atomic force microscopy (AFM) for a number of variables including duration of UV exposure, monomer concentration, and methacrylate functionalization. To determine how cells would respond to a specific two-step polymerization, NIH 3T3 fibroblasts were cultured on the stiffening MeHA hydrogels and found to reorganize their cytoskeleton and spread area upon hydrogel stiffening, consistent with cells originally cultured on substrates of the final elastic modulus.


Subject(s)
Biocompatible Materials/chemistry , Hyaluronic Acid/chemistry , Hydrogels/chemistry , Mechanical Phenomena , Methacrylates/chemistry , Animals , Biocompatible Materials/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Mice , NIH 3T3 Cells , Photochemical Processes , Polymerization , Ultraviolet Rays
3.
Adv Funct Mater ; 24(31): 4922-4926, 2014 Aug 20.
Article in English | MEDLINE | ID: mdl-26120293

ABSTRACT

The mechanical properties of the extracellular matrix (ECM) can dictate cell fate in biological systems. In tissue engineering, varying the stiffness of hydrogels-water-swollen polymeric networks that act as ECM substrates-has previously been demonstrated to control cell migration, proliferation, and differentiation. Here, "digital plasmonic patterning" (DPP) is developed to mechanically alter a hydrogel encapsulated with gold nanorods using a near-infrared laser, according to a digital (computer-generated) pattern. DPP can provide orders of magnitude changes in stiffness, and can be tuned by laser intensity and speed of writing. In vitro cellular experiments using A7R5 smooth muscle cells confirm cell migration and alignment according to these patterns, making DPP a useful technique for mechanically patterning hydrogels for various biomedical applications.

4.
Biomaterials ; 52: 140-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25818420

ABSTRACT

Polymerized high internal phase emulsion (polyHIPE) foams are extremely versatile materials for investigating cell-substrate interactions in vitro. Foam morphologies can be controlled by polymerization conditions to result in either open or closed pore structures with different levels of connectivity, consequently enabling the comparison between 2D and 3D matrices using the same substrate with identical surface chemistry conditions. Additionally, here we achieve the control of pore surface topology (i.e. how different ligands are clustered together) using amphiphilic block copolymers as emulsion stabilizers. We demonstrate that adhesion of human mesenchymal progenitor (hES-MP) cells cultured on polyHIPE foams is dependent on foam surface topology and chemistry but is independent of porosity and interconnectivity. We also demonstrate that the interconnectivity, architecture and surface topology of the foams has an effect on the osteogenic differentiation potential of hES-MP cells. Together these data demonstrate that the adhesive heterogeneity of a 3D scaffold could regulate not only mesenchymal stem cell attachment but also cell behavior in the absence of soluble growth factors.


Subject(s)
Biocompatible Materials/chemistry , Mesenchymal Stem Cells/cytology , Tissue Scaffolds/chemistry , Cell Adhesion , Cell Differentiation , Cell Line , Cell Proliferation , Humans , Mesenchymal Stem Cells/metabolism , Osteogenesis , Polymers/chemistry , Porosity
5.
Sci Rep ; 4: 6425, 2014 Sep 19.
Article in English | MEDLINE | ID: mdl-25236849

ABSTRACT

Cells secrete and assemble extracellular matrix throughout development, giving rise to time-dependent, tissue-specific stiffness. Mimicking myocardial matrix stiffening, i.e. ~10-fold increase over 1 week, with a hydrogel system enhances myofibrillar organization of embryonic cardiomyocytes compared to static hydrogels, and thus we sought to identify specific mechanosensitive proteins involved. Expression and/or phosphorylation state of 309 unique protein kinases were examined in embryonic cardiomyocytes plated on either dynamically stiffening or static mature myocardial stiffness hydrogels. Gene ontology analysis of these kinases identified cardiogenic pathways that exhibited time-dependent up-regulation on dynamic versus static matrices, including PI3K/AKT and p38 MAPK, while GSK3ß, a known antagonist of cardiomyocyte maturation, was down-regulated. Additionally, inhibiting GSK3ß on static matrices improved spontaneous contraction and myofibril organization, while inhibiting agonist AKT on dynamic matrices reduced myofibril organization and spontaneous contraction, confirming its role in mechanically-driven maturation. Together, these data indicate that mechanically-driven maturation is at least partially achieved via active mechanosensing at focal adhesions, affecting expression and phosphorylation of a variety of protein kinases important to cardiomyogenesis.


Subject(s)
MAP Kinase Signaling System/genetics , Myocytes, Cardiac/metabolism , Protein Kinases/biosynthesis , Vascular Stiffness/genetics , Animals , Cell Communication/genetics , Chick Embryo , Gene Expression Regulation , Myocytes, Cardiac/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction , Tissue Array Analysis
SELECTION OF CITATIONS
SEARCH DETAIL