Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Nov 11.
Article in English | MEDLINE | ID: mdl-37986921

ABSTRACT

The cell nucleus is continuously exposed to external signals, of both chemical and mechanical nature. To ensure proper cellular response, cells need to regulate not only the transmission of these signals, but also their timing and duration. Such timescale regulation is well described for fluctuating chemical signals, but if and how it applies to mechanical signals reaching the nucleus is still unknown. Here we demonstrate that the formation of fibrillar adhesions locks the nucleus in a mechanically deformed conformation, setting the mechanical response timescale to that of fibrillar adhesion remodelling (~1 hour). This process encompasses both mechanical deformation and associated mechanotransduction (such as via YAP), in response to both increased and decreased mechanical stimulation. The underlying mechanism is the anchoring of the vimentin cytoskeleton to fibrillar adhesions and the extracellular matrix through plectin 1f, which maintains nuclear deformation. Our results reveal a mechanism to regulate the timescale of mechanical adaptation, effectively setting a low pass filter to mechanotransduction.

2.
Polymers (Basel) ; 15(7)2023 Mar 28.
Article in English | MEDLINE | ID: mdl-37050285

ABSTRACT

This study describes the synthesis of novel amphiphilic linear-dendritic block copolymers and their self-assembly in water to form supramolecular nanoreactors capable of catalyzing Suzuki-Miyaura coupling reactions under "green" conditions. The block copolymers were formed through copper(I)-catalyzed alkyne-azide cycloaddition between azide functionalized poly(benzyl ether) dendrons as the perfectly branched blocks, as well as bis-alkyne modified poly(ethylene glycol), PEG, as the linear block. A first-generation poly(benzyl ether) dendron (G1) was coupled to a bis-alkyne modified PEG with molecular mass of 5 kDa, forming an ABA copolymer (G1)2-PEG5k-(G1)2 (yield 62%), while a second-generation dendron (G2) was coupled to a 11 kDa bis-alkyne modified PEG to produce (G2)2-PEG11k-(G2)2 (yield 49%). The structural purity and low dispersity of the linear-dendritic copolymers were verified by size-exclusion chromatography and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Their self-assembly was studied by dynamic light scattering, showing that (G1)2-PEG5k-(G1)2 and (G2)2-PEG11k-(G2)2 formed single populations of micelles (17 nm and 37 nm in diameter, respectively). The triazole rings located at the boundaries between the core and the corona are efficient chelating groups for transition metals. The ability of the micelles to complex Pd was confirmed by 1H NMR, transmission electron microscopy, and inductively coupled plasma. The catalytic activity of the supramolecular linear-dendritic/Pd complexes was tested in water by model Suzuki-Miyaura reactions in which quantitative yields were achieved within 3 h at 40 °C, while, at 17 °C, a yield of more than 70% was attained after 17 h.

3.
Sci Adv ; 9(3): eadd5668, 2023 01 20.
Article in English | MEDLINE | ID: mdl-36662859

ABSTRACT

Spatiotemporally coordinated transformations in epithelial curvature are necessary to generate crypt-villus structures during intestinal development. However, the temporal regulation of mechanotransduction pathways that drive crypt morphogenesis remains understudied. Intestinal organoids have proven useful to study crypt morphogenesis in vitro, yet the reliance on static culture scaffolds limits the ability to assess the temporal effects of changing curvature. Here, a photoinduced hydrogel cross-link exchange reaction is used to spatiotemporally alter epithelial curvature and study how dynamic changes in curvature influence mechanotransduction pathways to instruct crypt morphogenesis. Photopatterned curvature increased membrane tension and depolarization, which was required for subsequent nuclear localization of yes-associated protein 1 (YAP) observed 24 hours following curvature change. Curvature-directed crypt morphogenesis only occurred following a delay in the induction of differentiation that coincided with the delay in spatially restricted YAP localization, indicating that dynamic changes in curvature initiate epithelial curvature-dependent mechanotransduction pathways that temporally regulate crypt morphogenesis.


Subject(s)
Intestines , Mechanotransduction, Cellular , Intestinal Mucosa/metabolism , Organoids , Morphogenesis
4.
Sci Robot ; 8(74): eadc9800, 2023 01 25.
Article in English | MEDLINE | ID: mdl-36696474

ABSTRACT

Stiffness and forces are two fundamental quantities essential to living cells and tissues. However, it has been a challenge to quantify both 3D traction forces and stiffness (or modulus) using the same probe in vivo. Here, we describe an approach that overcomes this challenge by creating a magnetic microrobot probe with controllable functionality. Biocompatible ferromagnetic cobalt-platinum microcrosses were fabricated, and each microcross (about 30 micrometers) was trapped inside an arginine-glycine-apartic acid-conjugated stiff poly(ethylene glycol) (PEG) round microgel (about 50 micrometers) using a microfluidic device. The stiff magnetic microrobot was seeded inside a cell colony and acted as a stiffness probe by rigidly rotating in response to an oscillatory magnetic field. Then, brief episodes of ultraviolet light exposure were applied to dynamically photodegrade and soften the fluorescent nanoparticle-embedded PEG microgel, whose deformation and 3D traction forces were quantified. Using the microrobot probe, we show that malignant tumor-repopulating cell colonies altered their modulus but not traction forces in response to different 3D substrate elasticities. Stiffness and 3D traction forces were measured, and both normal and shear traction force oscillations were observed in zebrafish embryos from blastula to gastrula. Mouse embryos generated larger tensile and compressive traction force oscillations than shear traction force oscillations during blastocyst. The microrobot probe with controllable functionality via magnetic fields could potentially be useful for studying the mechanoregulation of cells, tissues, and embryos.


Subject(s)
Microgels , Neoplasms , Robotics , Animals , Mice , Zebrafish , Magnetic Phenomena
5.
Cell Stem Cell ; 29(5): 678-691, 2022 05 05.
Article in English | MEDLINE | ID: mdl-35413278

ABSTRACT

The recapitulation of complex microenvironments that regulate cell behavior during development, disease, and wound healing is key to understanding fundamental biological processes. In vitro, multicellular morphogenesis, organoid maturation, and disease modeling have traditionally been studied using either non-physiological 2D substrates or 3D biological matrices, neither of which replicate the spatiotemporal biochemical and biophysical complexity of biology. Here, we provide a guided overview of the recent advances in the programming of synthetic hydrogels that offer precise control over the spatiotemporal properties within cellular microenvironments, such as advances in the control of cell-driven remodeling, bioprinting, or user-defined manipulation of properties (e.g., via light irradiation).


Subject(s)
Bioprinting , Hydrogels , Cellular Microenvironment , Hydrogels/chemistry , Organoids , Tissue Engineering , Wound Healing
6.
ACS Biomater Sci Eng ; 8(11): 4634-4638, 2022 Nov 14.
Article in English | MEDLINE | ID: mdl-35298149

ABSTRACT

Intestinal organoids are self-organized tissue constructs, grown in vitro, that resemble the structure and function of the intestine and are often considered promising as a prospective platform for drug testing and disease modeling. Organoid development in vitro is typically instructed by exogenous cues delivered from the media, but cellular responses also depend on properties of the surrounding microenvironmental niche, such as mechanical stiffness and extracellular matrix (ECM) ligands. In recent years, synthetic hydrogel platforms have been engineered to resemble the in vivo niche, with the goal of generating physiologically relevant environments that can promote mature and reproducible organoid development. However, a few of these approaches consider the importance of intestinal organoid morphology or how morphology changes during development, as cues that may dictate organoid functionality. For example, intestinal organoids grown in vitro often lack the physical boundary conditions found in vivo that are responsible for shaping a collection of cells into developmentally relevant morphologies, resulting in organoids that often differ in structure and cellular organization from the parent organ. This disconnect relates, in part, to a lack of appropriate adaptable and programmable materials for cell culture, especially those that enable control over colony growth and differentiation in space and time (i.e., 4D materials). We posit that the future of organoid culture platforms may benefit from advances in photoadaptable chemistries and integration into biomaterials scaffolds, thereby allowing greater user-directed control over both the macro- and microscale material properties. In this way, synthetic materials can begin to better replicate changes in the ECM during development or regeneration in vivo. Recapitulation of cellular and tissue morphological changes, along with an appreciation for the appropriate developmental time scales, should help instruct the next generation of organoid models to facilitate predictable outcomes.


Subject(s)
Cell Culture Techniques , Organoids , Organoids/physiology , Intestines , Biocompatible Materials , Extracellular Matrix/chemistry
7.
Adv Mater ; 34(16): e2109252, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35182403

ABSTRACT

3D organoid models have recently seen a boom in popularity, as they can better recapitulate the complexity of multicellular organs compared to other in vitro culture systems. However, organoids are difficult to image because of the limited penetration depth of high-resolution microscopes and depth-dependent light attenuation, which can limit the understanding of signal transduction pathways and characterization of intimate cell-extracellular matrix (ECM) interactions. To overcome these challenges, phototransfer by allyl sulfide exchange-expansion microscopy (PhASE-ExM) is developed, enabling optical clearance and super-resolution imaging of organoids and their ECM in 3D. PhASE-ExM uses hydrogels prepared via photoinitiated polymerization, which is advantageous as it decouples monomer diffusion into thick organoid cultures from the hydrogel fabrication. Apart from compatibility with organoids cultured in Matrigel, PhASE-ExM enables 3.25× expansion and super-resolution imaging of organoids cultured in synthetic poly(ethylene glycol) (PEG) hydrogels crosslinked via allyl-sulfide groups (PEG-AlS) through simultaneous photopolymerization and radical-mediated chain-transfer reactions that complete in <70 s. Further, PEG-AlS hydrogels can be in situ softened to promote organoid crypt formation, providing a super-resolution imaging platform both for pre- and post-differentiated organoids. Overall, PhASE-ExM is a useful tool to decipher organoid behavior by enabling sub-micrometer scale, 3D visualization of proteins and signal transduction pathways.


Subject(s)
Microscopy , Organoids , Allyl Compounds , Biocompatible Materials/metabolism , Extracellular Matrix , Hydrogels/metabolism , Organoids/metabolism , Sulfides
8.
Biofabrication ; 13(4)2021 09 02.
Article in English | MEDLINE | ID: mdl-34380115

ABSTRACT

Biofabrication allows for the templating of structural features in materials on cellularly-relevant size scales, enabling the generation of tissue-like structures with controlled form and function. This is particularly relevant for growing organoids, where the application of biochemical and biomechanical stimuli can be used to guide the assembly and differentiation of stem cells and form architectures similar to the parent tissue or organ. Recently, ablative laser-scanning techniques was used to create 3D overhang features in collagen hydrogels at size scales of 10-100µm and supported the crypt-villus architecture in intestinal organoids. As a complementary method, providing advantages for high-throughput patterning, we printed thioester functionalized poly(ethylene glycol) (PEG) elastomers using digital light processing (DLP) and created sacrificial, 3D shapes that could be molded into soft (G' < 1000 Pa) hydrogel substrates. Specifically, three-arm 1.3 kDa PEG thiol and three-arm 1.6 kDa PEG norbornene, containing internal thioester groups, were photopolymerized to yield degradable elastomers. When incubated in a solution of 300 mM 2-mercaptoethanol (pH 9.0), 1 mm thick 10 mm diameter elastomer discs degraded in <2 h. Using DLP, arrays of features with critical dimensions of 37 ± 4µm, resolutions of 22 ± 5µm, and overhang structures as small as 50µm, were printed on the order of minutes. These sacrificial thioester molds with physiologically relevant features were cast-molded into Matrigel and subsequently degraded to create patterned void spaces with high fidelity. Intestinal stem cells (ISCs) cultured on the patterned Matrigel matrices formed confluent monolayers that conformed to the underlying pattern. DLP printed sacrificial thioester elastomer constructs provide a robust and rapid method to fabricate arrays of 3D organoid-sized features in soft tissue culture substrates and should enable investigations into the effect of epithelial geometry and spacing on the growth and differentiation of ISCs.


Subject(s)
Elastomers , Organoids , Hydrogels , Polyethylene Glycols , Printing, Three-Dimensional
9.
Arterioscler Thromb Vasc Biol ; 40(11): e296-e308, 2020 11.
Article in English | MEDLINE | ID: mdl-32938214

ABSTRACT

OBJECTIVE: Resident valvular interstitial cells (VICs) activate to myofibroblasts during aortic valve stenosis progression, which further promotes fibrosis or even differentiate into osteoblast-like cells that can lead to calcification of valve tissue. Inflammation is a hallmark of aortic valve stenosis, so we aimed to determine proinflammatory cytokines secreted from M1 macrophages that give rise to a transient VIC phenotype that leads to calcification of valve tissue. Approach and Results: We designed hydrogel biomaterials as valve extracellular matrix mimics enabling the culture of VICs in either their quiescent fibroblast or activated myofibroblast phenotype in response to the local matrix stiffness. When VIC fibroblasts and myofibroblasts were treated with conditioned media from THP-1-derived M1 macrophages, we observed robust reduction of αSMA (alpha smooth muscle actin) expression, reduced stress fiber formation, and increased proliferation, suggesting a potent antifibrotic effect. We further identified TNF (tumor necrosis factor)-α and IL (interleukin)-1ß as 2 cytokines in M1 media that cause the observed antifibrotic effect. After 7 days of culture in M1 conditioned media, VICs began differentiating into osteoblast-like cells, as measured by increased expression of RUNX2 (runt-related transcription factor 2) and osteopontin. We also identified and validated IL-6 as a critical mediator of the observed pro-osteogenic effect. CONCLUSIONS: Proinflammatory cytokines in M1 conditioned media inhibit myofibroblast activation in VICs (eg, TNF-α and IL-1ß) and promote their osteogenic differentiation (eg, IL-6). Together, our work suggests inflammatory M1 macrophages may drive a myofibroblast-to-osteogenic intermediate VIC phenotype, which may mediate the switch from fibrosis to calcification during aortic valve stenosis progression.


Subject(s)
Aortic Valve Stenosis/metabolism , Aortic Valve/metabolism , Aortic Valve/pathology , Calcinosis/metabolism , Cytokines/metabolism , Inflammation Mediators/metabolism , Macrophages/metabolism , Myofibroblasts/metabolism , Osteoblasts/metabolism , Osteogenesis , Paracrine Communication , Animals , Aortic Valve Stenosis/pathology , Calcinosis/pathology , Cell Proliferation , Extracellular Matrix/metabolism , Fibrosis , Humans , Male , Myofibroblasts/pathology , Osteoblasts/pathology , Phenotype , Secretory Pathway , Signal Transduction , Sus scrofa , THP-1 Cells
10.
Adv Mater ; 32(30): e1905366, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32548863

ABSTRACT

Intestinal organoids are useful in vitro models for basic and translational studies aimed at understanding and treating disease. However, their routine culture relies on animal-derived matrices that limit translation to clinical applications. In fact, there are few fully defined, synthetic hydrogel systems that allow for the expansion of intestinal organoids. Here, an allyl sulfide photodegradable hydrogel is presented, achieving rapid degradation through radical addition-fragmentation chain transfer (AFCT) reactions, to support routine passaging of intestinal organoids. Shear rheology to first characterize the effect of thiol and allyl sulfide crosslink structures on degradation kinetics is used. Irradiation with 365 nm light (5 mW cm-2 ) in the presence of a soluble thiol (glutathione at 15 × 10-3 m), and a photoinitiator (lithium phenyl-2,4,6-trimethylbenzoylphosphinate at 1 × 10-3 m), leads to complete hydrogel degradation in less than 15 s. Allyl sulfide hydrogels are used to support the formation of epithelial colonies from single intestinal stem cells, and rapid photodegradation is used to achieve repetitive passaging of stem cell colonies without loss in morphology or organoid formation potential. This platform could support long-term culture of intestinal organoids, potentially replacing the need for animal-derived matrices, while also allowing systematic variations to the hydrogel properties tailored for the organoid of interest.


Subject(s)
Allyl Compounds/chemistry , Hydrogels/chemistry , Hydrogels/metabolism , Organoids/metabolism , Photolysis , Sulfhydryl Compounds/chemistry , Sulfides/chemistry , Animals , Intestinal Mucosa/cytology , Light , Mice , Rheology , Shear Strength , Solubility
11.
Adv Healthc Mater ; 9(8): e1901214, 2020 04.
Article in English | MEDLINE | ID: mdl-31957249

ABSTRACT

Intestinal organoid protocols rely on the use of extracellular scaffolds, typically Matrigel, and upon switching from growth to differentiation promoting media, a symmetry breaking event takes place. During this stage, the first bud like structures analogous to crypts protrude from the central body and differentiation ensues. While organoids provide unparalleled architectural and functional complexity, this sophistication is also responsible for the high variability and lack of reproducibility of uniform crypt-villus structures. If function follows form in organoids, such structural variability carries potential limitations for translational applications (e.g., drug screening). Consequently, there is interest in developing synthetic biomaterials to direct organoid growth and differentiation. It has been hypothesized that synthetic scaffold softening is necessary for crypt development, and these mechanical requirements raise the question, what compressive forces and subsequent relaxation are necessary for organoid maturation? To that end, allyl sulfide hydrogels are employed as a synthetic extracellular matrix mimic, but with photocleavable bonds that temporally regulate the material's bulk modulus. By varying the extent of matrix softening, it is demonstrated that crypt formation, size, and number per colony are functions of matrix softening. An understanding of the mechanical dependence of crypt architecture is necessary to instruct homogenous, reproducible organoids for clinical applications.


Subject(s)
Intestines , Organoids , Extracellular Matrix , Intestinal Mucosa , Reproducibility of Results
12.
J Am Chem Soc ; 140(37): 11585-11588, 2018 09 19.
Article in English | MEDLINE | ID: mdl-30183266

ABSTRACT

Muscle cells sense the mechanical properties of their microenvironment, and these properties can change in response to injury or disease. Hydrogels with dynamic material properties can be used to study the effect of such varying mechanical signals. Here, we report the ability of azadibenzocyclooctyne to undergo a cytocompatible, photoinitiated crosslinking reaction. This reaction is exploited as a strategy for on-demand stiffening of three-dimensional cell scaffolds formed through an initial strain-promoted azide-alkyne cycloaddition. Myoblasts encapsulated in these networks respond to increased matrix stiffness through decreased cell spreading and nuclear localization of Yes-associated protein 1 (YAP). However, when the photocrosslinking reaction is delayed to allow cell spreading, elongated myoblasts display increased YAP nuclear localization.


Subject(s)
Aza Compounds/chemistry , Cross-Linking Reagents/chemistry , Cyclooctanes/chemistry , Hydrogels/chemistry , Mechanotransduction, Cellular , Myoblasts/cytology , Cell Survival , Humans , Molecular Structure , Photochemical Processes
SELECTION OF CITATIONS
SEARCH DETAIL
...