Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.187
Filter
Add more filters

Publication year range
1.
Annu Rev Biochem ; 87: 351-390, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29195049

ABSTRACT

In this review, we describe speculative ideas and early stage research concerning the flow of genetic information from the nuclear residence of genes to the disparate, cytoplasmic sites of protein synthesis. We propose that this process of information transfer is meticulously guided by transient structures formed from protein segments of low sequence complexity/intrinsic disorder. These low complexity domains are ubiquitously associated with regulatory proteins that control gene expression and RNA biogenesis, but they are also found in the central channel of nuclear pores, the nexus points of intermediate filament assembly, and the locations of action of other well-studied cellular proteins and pathways. Upon being organized into localized cellular positions via mechanisms utilizing properly folded protein domains, thereby facilitating elevated local concentration, certain low complexity domains adopt cross-ß interactions that are both structurally specific and labile to disassembly. These weakly tethered assemblies, we propose, are built to relay the passage of genetic information from one site to another within a cell, ensuring that the process is of extreme fidelity.


Subject(s)
Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/metabolism , Models, Biological , Animals , Cytoplasmic Granules/genetics , Cytoplasmic Granules/metabolism , Gene Expression , Heredodegenerative Disorders, Nervous System/genetics , Heredodegenerative Disorders, Nervous System/metabolism , Humans , Hydrogels , Intrinsically Disordered Proteins/chemistry , Models, Molecular , Mutation , Protein Interaction Domains and Motifs , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Protein FUS/chemistry , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism
2.
Proc Natl Acad Sci U S A ; 121(23): e2316206121, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38805271

ABSTRACT

Rapid progress in algal biotechnology has triggered a growing interest in hydrogel-encapsulated microalgal cultivation, especially for the engineering of functional photosynthetic materials and biomass production. An overlooked characteristic of gel-encapsulated cultures is the emergence of cell aggregates, which are the result of the mechanical confinement of the cells. Such aggregates have a dramatic effect on the light management of gel-encapsulated photobioreactors and hence strongly affect the photosynthetic outcome. To evaluate such an effect, we experimentally studied the optical response of hydrogels containing algal aggregates and developed optical simulations to study the resultant light intensity profiles. The simulations are validated experimentally via transmittance measurements using an integrating sphere and aggregate volume analysis with confocal microscopy. Specifically, the heterogeneous distribution of cell aggregates in a hydrogel matrix can increase light penetration while alleviating photoinhibition more effectively than in a flat biofilm. Finally, we demonstrate that light harvesting efficiency can be further enhanced with the introduction of scattering particles within the hydrogel matrix, leading to a fourfold increase in biomass growth. Our study, therefore, highlights a strategy for the design of spatially efficient photosynthetic living materials that have important implications for the engineering of future algal cultivation systems.


Subject(s)
Hydrogels , Light , Microalgae , Photosynthesis , Hydrogels/chemistry , Microalgae/growth & development , Microalgae/metabolism , Biomass , Photobioreactors
3.
Proc Natl Acad Sci U S A ; 121(35): e2406787121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39163337

ABSTRACT

Muscle stem cells (MuSCs) are specialized cells that reside in adult skeletal muscle poised to repair muscle tissue. The ability of MuSCs to regenerate damaged tissues declines markedly with aging and in diseases such as Duchenne muscular dystrophy, but the underlying causes of MuSC dysfunction remain poorly understood. Both aging and disease result in dramatic increases in the stiffness of the muscle tissue microenvironment from fibrosis. MuSCs are known to lose their regenerative potential if cultured on stiff plastic substrates. We sought to determine whether MuSCs harbor a memory of their past microenvironment and if it can be overcome. We tested MuSCs in situ using dynamic hydrogel biomaterials that soften or stiffen on demand in response to light and found that freshly isolated MuSCs develop a persistent memory of substrate stiffness characterized by loss of proliferative progenitors within the first three days of culture on stiff substrates. MuSCs cultured on soft hydrogels had altered cytoskeletal organization and activity of Rho and Rac guanosine triphosphate hydrolase (GTPase) and Yes-associated protein mechanotransduction pathways compared to those on stiff hydrogels. Pharmacologic inhibition identified RhoA activation as responsible for the mechanical memory phenotype, and single-cell RNA sequencing revealed a molecular signature of the mechanical memory. These studies highlight that microenvironmental stiffness regulates MuSC fate and leads to MuSC dysfunction that is not readily reversed by changing stiffness. Our results suggest that stiffness can be circumvented by targeting downstream signaling pathways to overcome stem cell dysfunction in aged and disease states with aberrant fibrotic tissue mechanics.


Subject(s)
Biocompatible Materials , Hydrogels , Muscle, Skeletal , Animals , Hydrogels/chemistry , Biocompatible Materials/chemistry , Muscle, Skeletal/metabolism , Mice , Mechanotransduction, Cellular , Stem Cells/metabolism , Stem Cells/cytology , rhoA GTP-Binding Protein/metabolism , Cells, Cultured
4.
Proc Natl Acad Sci U S A ; 121(6): e2309457121, 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38289949

ABSTRACT

Relating the macroscopic properties of protein-based materials to their underlying component microstructure is an outstanding challenge. Here, we exploit computational design to specify the size, flexibility, and valency of de novo protein building blocks, as well as the interaction dynamics between them, to investigate how molecular parameters govern the macroscopic viscoelasticity of the resultant protein hydrogels. We construct gel systems from pairs of symmetric protein homo-oligomers, each comprising 2, 5, 24, or 120 individual protein components, that are crosslinked either physically or covalently into idealized step-growth biopolymer networks. Through rheological assessment, we find that the covalent linkage of multifunctional precursors yields hydrogels whose viscoelasticity depends on the crosslink length between the constituent building blocks. In contrast, reversibly crosslinking the homo-oligomeric components with a computationally designed heterodimer results in viscoelastic biomaterials exhibiting fluid-like properties under rest and low shear, but solid-like behavior at higher frequencies. Exploiting the unique genetic encodability of these materials, we demonstrate the assembly of protein networks within living mammalian cells and show via fluorescence recovery after photobleaching (FRAP) that mechanical properties can be tuned intracellularly in a manner similar to formulations formed extracellularly. We anticipate that the ability to modularly construct and systematically program the viscoelastic properties of designer protein-based materials could have broad utility in biomedicine, with applications in tissue engineering, therapeutic delivery, and synthetic biology.


Subject(s)
Biocompatible Materials , Hydrogels , Animals , Hydrogels/chemistry , Biopolymers , Mammals
5.
Proc Natl Acad Sci U S A ; 121(19): e2322822121, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38687784

ABSTRACT

Hydrogels derived from decellularized extracellular matrices (ECM) of animal origin show immense potential for regenerative applications due to their excellent cytocompatibility and biomimetic properties. Despite these benefits, the impact of decellularization protocols on the properties and immunogenicity of these hydrogels remains relatively unexplored. In this study, porcine skeletal muscle ECM (smECM) underwent decellularization using mechanical disruption (MD) and two commonly employed decellularization detergents, sodium deoxycholate (SDC) or Triton X-100. To mitigate immunogenicity associated with animal-derived ECM, all decellularized tissues were enzymatically treated with α-galactosidase to cleave the primary xenoantigen-the α-Gal antigen. Subsequently, the impact of the different decellularization protocols on the resultant hydrogels was thoroughly investigated. All methods significantly reduced total DNA content in hydrogels. Moreover, α-galactosidase treatment was crucial for cleaving α-Gal antigens, suggesting that conventional decellularization methods alone are insufficient. MD preserved total protein, collagen, sulfated glycosaminoglycan, laminin, fibronectin, and growth factors more efficiently than other protocols. The decellularization method impacted hydrogel gelation kinetics and ultrastructure, as confirmed by turbidimetric and scanning electron microscopy analyses. MD hydrogels demonstrated high cytocompatibility, supporting satellite stem cell recruitment, growth, and differentiation into multinucleated myofibers. In contrast, the SDC and Triton X-100 protocols exhibited cytotoxicity. Comprehensive in vivo immunogenicity assessments in a subcutaneous xenotransplantation model revealed MD hydrogels' biocompatibility and low immunogenicity. These findings highlight the significant influence of the decellularization protocol on hydrogel properties. Our results suggest that combining MD with α-galactosidase treatment is an efficient method for preparing low-immunogenic smECM-derived hydrogels with enhanced properties for skeletal muscle regenerative engineering and clinical applications.


Subject(s)
Extracellular Matrix , Hydrogels , Muscle, Skeletal , Animals , Hydrogels/chemistry , Swine , Extracellular Matrix/metabolism , Tissue Engineering/methods , Decellularized Extracellular Matrix/chemistry , Mice , alpha-Galactosidase/immunology , alpha-Galactosidase/metabolism , Deoxycholic Acid/chemistry , Octoxynol/chemistry
6.
Development ; 150(13)2023 07 01.
Article in English | MEDLINE | ID: mdl-37387575

ABSTRACT

The development of multicellular complex organisms relies on coordinated signaling from the microenvironment, including both biochemical and mechanical interactions. To better understand developmental biology, increasingly sophisticated in vitro systems are needed to mimic these complex extracellular features. In this Primer, we explore how engineered hydrogels can serve as in vitro culture platforms to present such signals in a controlled manner and include examples of how they have been used to advance our understanding of developmental biology.


Subject(s)
Hydrogels , Signal Transduction
7.
Proc Natl Acad Sci U S A ; 120(51): e2220755120, 2023 Dec 19.
Article in English | MEDLINE | ID: mdl-38091296

ABSTRACT

Fibrous networks formed by biological polymers such as collagen or fibrin exhibit nonlinear mechanical behavior. They undergo strong stiffening in response to weak shear and elongational strains, but soften under compressional strain, in striking difference with the response to the deformation of flexible-strand networks formed by molecules. The nonlinear properties of fibrous networks are attributed to the mechanical asymmetry of the constituent filaments, for which a stretching modulus is significantly larger than the bending modulus. Studies of the nonlinear mechanical behavior are generally performed on hydrogels formed by biological polymers, which offers limited control over network architecture. Here, we report an engineered covalently cross-linked nanofibrillar hydrogel derived from cellulose nanocrystals and gelatin. The variation in hydrogel composition provided a broad-range change in its shear modulus. The hydrogel exhibited both shear-stiffening and compression-induced softening, in agreement with the predictions of the affine model. The threshold nonlinear stress and strain were universal for the hydrogels with different compositions, which suggested that nonlinear mechanical properties are general for networks formed by rigid filaments. The experimental results were in agreement with an affine model describing deformation of the network formed by rigid filaments. Our results lend insight into the structural features that govern the nonlinear biomechanics of fibrous networks and provide a platform for future studies of the biological impact of nonlinear mechanical properties.


Subject(s)
Collagen , Hydrogels , Hydrogels/chemistry , Collagen/chemistry , Elasticity , Polymers , Cytoskeleton , Stress, Mechanical
8.
Proc Natl Acad Sci U S A ; 120(35): e2307772120, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37603747

ABSTRACT

Artificial cells are biomimetic structures formed from molecular building blocks that replicate biological processes, behaviors, and architectures. Of these building blocks, hydrogels have emerged as ideal, yet underutilized candidates to provide a gel-like chassis in which to incorporate both biological and nonbiological componentry which enables the replication of cellular functionality. Here, we demonstrate a microfluidic strategy to assemble biocompatible cell-sized hydrogel-based artificial cells with a variety of different embedded functional subcompartments, which act as engineered synthetic organelles. The organelles enable the recreation of increasingly biomimetic behaviors, including stimulus-induced motility, content release through activation of membrane-associated proteins, and enzymatic communication with surrounding bioinspired compartments. In this way, we showcase a foundational strategy for the bottom-up construction of hydrogel-based artificial cell microsystems which replicate fundamental cellular behaviors, paving the way for the construction of next-generation biotechnological devices.


Subject(s)
Artificial Cells , Biomimetics , Hydrogels , Communication , Organelles
9.
Proc Natl Acad Sci U S A ; 120(8): e2213030120, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36791112

ABSTRACT

Load-bearing soft tissues normally show J-shaped stress-strain behaviors with high compliance at low strains yet high strength at high strains. They have high water content but are still tough and durable. By contrast, naturally derived hydrogels are weak and brittle. Although hydrogels prepared from synthetic polymers can be strong and tough, they do not have the desired bioactivity for emerging biomedical applications. Here, we present a thermomechanical approach to replicate the combinational properties of soft tissues in protein-based photocrosslinkable hydrogels. As a demonstration, we create a gelatin methacryloyl fiber hydrogel with soft tissue-like mechanical properties, such as low Young's modulus (0.1 to 0.3 MPa), high strength (1.1 ± 0.2 MPa), high toughness (9,100 ± 2,200 J/m3), and high fatigue resistance (2,300 ± 500 J/m2). This hydrogel also resembles the biochemical and architectural properties of native extracellular matrix, which enables a fast formation of 3D interconnected cell meshwork inside hydrogels. The fiber architecture also regulates cellular mechanoresponse and supports cell remodeling inside hydrogels. The integration of tissue-like mechanical properties and bioactivity is highly desirable for the next-generation biomaterials and could advance emerging fields such as tissue engineering and regenerative medicine.


Subject(s)
Biocompatible Materials , Hydrogels , Hydrogels/chemistry , Biocompatible Materials/chemistry , Tissue Engineering , Water/chemistry , Polymers
10.
Proc Natl Acad Sci U S A ; 120(17): e2220565120, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37071684

ABSTRACT

DNA-based biomaterials have been proposed for tissue engineering approaches due to their predictable assembly into complex morphologies and ease of functionalization. For bone tissue regeneration, the ability to bind Ca2+ and promote hydroxyapatite (HAP) growth along the DNA backbone combined with their degradation and release of extracellular phosphate, a known promoter of osteogenic differentiation, make DNA-based biomaterials unlike other currently used materials. However, their use as biodegradable scaffolds for bone repair remains scarce. Here, we describe the design and synthesis of DNA hydrogels, gels composed of DNA that swell in water, their interactions in vitro with the osteogenic cell lines MC3T3-E1 and mouse calvarial osteoblast, and their promotion of new bone formation in rat calvarial wounds. We found that DNA hydrogels can be readily synthesized at room temperature, and they promote HAP growth in vitro, as characterized by Fourier transform infrared spectroscopy, X-ray diffraction, scanning electron microscopy, atomic force microscopy, and transmission electron microscopy. Osteogenic cells remain viable when seeded on DNA hydrogels in vitro, as characterized by fluorescence microscopy. In vivo, DNA hydrogels promote the formation of new bone in rat calvarial critical size defects, as characterized by micro-computed tomography and histology. This study uses DNA hydrogels as a potential therapeutic biomaterial for regenerating lost bone.


Subject(s)
Hydrogels , Osteogenesis , Mice , Rats , Animals , Hydrogels/chemistry , X-Ray Microtomography , Bone Regeneration , Biocompatible Materials/pharmacology , Biocompatible Materials/chemistry , Durapatite/pharmacology , Durapatite/chemistry , Tissue Engineering , Tissue Scaffolds/chemistry
11.
Semin Cell Dev Biol ; 147: 58-69, 2023 09 30.
Article in English | MEDLINE | ID: mdl-36732105

ABSTRACT

Scientific knowledge in the field of cell biology and mechanobiology heavily leans on cell-based in vitro experiments and models that favor the examination and comprehension of certain biological processes and occurrences across a variety of environments. Cell culture assays are an invaluable instrument for a vast spectrum of biomedical and biophysical investigations. The quality of experimental models in terms of simplicity, reproducibility, and combinability with other methods, and in particular the scale at which they depict cell fate in native tissues, is critical to advancing the knowledge of the comprehension of cell-cell and cell-matrix interactions in tissues and organs. Typically, in vitro models are centered on the experimental tinkering of mammalian cells, most often cultured as monolayers on planar, two-dimensional (2D) materials. Notwithstanding the significant advances and numerous findings that have been accomplished with flat biology models, their usefulness for generating further new biological understanding is constrained because the simple 2D setting does not reproduce the physiological response of cells in natural living tissues. In addition, the co-culture systems in a 2D stetting weakly mirror their natural environment of tissues and organs. Significant advances in 3D cell biology and matrix engineering have resulted in the creation and establishment of a new type of cell culture shapes that more accurately represents the in vivo microenvironment and allows cells and their interactions to be analyzed in a biomimetic approach. Contemporary biomedical and biophysical science has novel advances in technology that permit the design of more challenging and resilient in vitro models for tissue engineering, with a particular focus on scaffold- or hydrogel-based formats, organotypic cultures, and organs-on-chips, which cover the purposes of co-cultures. Even these complex systems must be kept as simplified as possible in order to grasp a particular section of physiology too very precisely. In particular, it is highly appreciated that they bridge the space between conventional animal research and human (patho)physiology. In this review, the recent progress in 3D biomimetic culturation is presented with a special focus on co-cultures, with an emphasis on the technological building blocks and endothelium-based co-culture models in cancer research that are available for the development of more physiologically relevant in vitro models of human tissues under normal and diseased conditions. Through applications and samples of various physiological and disease models, it is possible to identify the frontiers and future engagement issues that will have to be tackled to integrate synthetic biomimetic culture systems far more successfully into biomedical and biophysical investigations.


Subject(s)
Cell Culture Techniques , Tissue Engineering , Animals , Humans , Coculture Techniques , Reproducibility of Results , Tissue Engineering/methods , Endothelial Cells , Mammals
12.
Methods ; 228: 55-64, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38782295

ABSTRACT

Metal ions, including biologically prevalent sodium ions, can modulate electrostatic interactions frequently involved in the stability of condensed compartments in cells. Quantitative characterization of heterogeneous ion dynamics inside biomolecular condensates demands new experimental approaches. Here we develop a 23Na NMR relaxation-based integrative approach to probe dynamics of sodium ions inside agarose gels as a model system. We exploit the electric quadrupole moment of spin-3/2 23Na nuclei and, through combination of single-quantum and triple-quantum-filtered 23Na NMR relaxation methods, disentangle the relaxation contribution of different populations of sodium ions inside gels. Three populations of sodium ions are identified: a population with bi-exponential relaxation representing ions within the slow motion regime and two populations with mono-exponential relaxation but at different rates. Our study demonstrates the dynamical heterogeneity of sodium ions inside agarose gels and presents a new experimental approach for monitoring dynamics of sodium and other spin-3/2 ions (e.g. chloride) in condensed environments.


Subject(s)
Gels , Sepharose , Sodium , Sepharose/chemistry , Sodium/chemistry , Gels/chemistry , Magnetic Resonance Spectroscopy/methods , Ions/chemistry , Quantum Theory
13.
Proc Natl Acad Sci U S A ; 119(44): e2208040119, 2022 11.
Article in English | MEDLINE | ID: mdl-36279452

ABSTRACT

Organoid technology has provided unique insights into human organ development, function, and diseases. Patient-derived organoids are increasingly used for drug screening, modeling rare disorders, designing regenerative therapies, and understanding disease pathogenesis. However, the use of Matrigel to grow organoids represents a major challenge in the clinical translation of organoid technology. Matrigel is a poorly defined mixture of extracellular matrix proteins and growth factors extracted from the Engelbreth-Holm-Swarm mouse tumor. The extracellular matrix is a major driver of multiple cellular processes and differs significantly between tissues as well as in healthy and disease states of the same tissue. Therefore, we envisioned that the extracellular matrix derived from a native healthy tissue would be able to support organoid growth akin to organogenesis in vivo. Here, we have developed hydrogels from decellularized human and bovine endometrium. These hydrogels supported the growth of mouse and human endometrial organoids, which was comparable to Matrigel. Organoids grown in endometrial hydrogels were proteomically more similar to the native tissue than those cultured in Matrigel. Proteomic and Raman microspectroscopy analyses showed that the method of decellularization affects the biochemical composition of hydrogels and, subsequently, their ability to support organoid growth. The amount of laminin in hydrogels correlated with the number and shape of organoids. We also demonstrated the utility of endometrial hydrogels in developing solid scaffolds for supporting high-throughput, cell culture-based applications. In summary, endometrial hydrogels overcome a major limitation of organoid technology and greatly expand the applicability of organoids to understand endometrial biology and associated pathologies.


Subject(s)
Neoplasms , Organoids , Female , Humans , Cattle , Animals , Organoids/metabolism , Hydrogels/chemistry , Laminin/pharmacology , Laminin/metabolism , Proteomics , Endometrium , Neoplasms/metabolism
14.
Proc Natl Acad Sci U S A ; 119(36): e2207422119, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36037384

ABSTRACT

Understanding the physical principle that governs the stimuli-induced swelling and shrinking kinetics of hydrogels is indispensable for their applications. Here, we show that the shrinking and swelling kinetics of self-healing hydrogels could be intrinsically asymmetric. The structure frustration, formed by the large difference in the heat and solvent diffusions, remarkably slows down the shrinking kinetics. The plateau modulus of viscoelastic gels is found to be a key parameter governing the formation of structure frustration and, in turn, the asymmetric swelling and shrinking kinetics. This work provides fundamental understandings on the temperature-triggered transient structure formation in self-healing hydrogels. Our findings will find broad use in diverse applications of self-healing hydrogels, where cooperative diffusion of water and gel network is involved. Our findings should also give insight into the molecular diffusion in biological systems that possess macromolecular crowding environments similar to self-healing hydrogels.


Subject(s)
Hydrogels , Temperature , Diffusion , Hydrogels/chemistry , Kinetics , Water/chemistry
15.
Proc Natl Acad Sci U S A ; 119(39): e2123156119, 2022 09 27.
Article in English | MEDLINE | ID: mdl-36122212

ABSTRACT

Straightforward manufacturing pathways toward large-scale, uniformly layered composites may enable the next generation of materials with advanced optical, thermal, and mechanical properties. Reaction-diffusion systems are attractive candidates to this aim, but while layered composites theoretically could spontaneously arise from reaction-diffusion, in practice randomly oriented patches separated by defects form, yielding nonuniformly patterned materials. A propagating reaction front can prevent such nonuniform patterning, as is the case for Liesegang processes, in which diffusion drives a reaction front to produce layered precipitation patterns. However, while diffusion is crucial to control patterning, it slows down transport of reactants to the front and results in a steady increase of the band spacing as the front advances. Here, we circumvent these diffusive limitations by embedding the Liesegang process in mechanically responsive hydrogels. The coupling between a moving reaction front and hydrogel contraction induces the formation of a self-regulated transport channel that ballistically carries reactants toward the area where patterning occurs. This ensures rapid and uniform patterning. Specifically, large-scale ([Formula: see text]5-cm) uniform banding patterns are produced with tunable band distance (d = 60 to 160 µm) of silver dichromate crystals inside responsive gelatin-alginate hydrogels. The generality and applicability of our mechanoreaction-diffusion strategy are demonstrated by forming patterns of precipitates in significantly smaller microscopic banding patterns (d = 10 to 30 µm) that act as self-organized diffraction gratings. By circumventing the inherent limitations of diffusion, our strategy unlocks the potential of reaction-diffusion processes for the manufacturing of uniformly layered materials.


Subject(s)
Hydrogels , Manufactured Materials , Alginates/chemistry , Chromates/chemistry , Diffusion , Gelatin/chemistry , Hydrogels/chemistry , Silver/chemistry
16.
Nano Lett ; 24(7): 2188-2195, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38324001

ABSTRACT

Over the past few decades, hydrogels have attracted considerable attention as promising biomedical materials. However, conventional hydrogels require improved mechanical properties, such as brittleness, which significantly limits their widespread use. Recently, hydrogels with remarkably improved toughness have been developed; however, their low biocompatibility must be addressed. In this study, we developed a tough graphene hybrid hydrogel with nanostructures. The resultant hydrogel exhibited remarkable mechanical properties while representing an aligned nanostructure that resembled the extracellular matrix of soft tissue. Owing to the synergistic effect of the topographical properties, and the enhanced biochemical properties, the graphene hybrid hydrogel had excellent stretchability, resilience, toughness, and biocompatibility. Furthermore, the hydrogel displayed outstanding tissue regeneration capabilities (e.g., skin and tendons). Overall, the proposed graphene hybrid tough hydrogel may provide significant insights into the application of tough hydrogels in tissue regeneration.


Subject(s)
Graphite , Nanostructures , Hydrogels/chemistry , Graphite/chemistry , Biocompatible Materials/chemistry , Nanostructures/therapeutic use
17.
Nano Lett ; 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38591912

ABSTRACT

Deviations between macrorheological and particle-based microrheological measurements are often considered to be a nuisance and neglected. We study aqueous poly(ethylene oxide) (PEO) hydrogels for varying PEO concentrations and chain lengths that contain microscopic tracer particles and show that these deviations reveal the nanoscopic viscoelastic properties of the particle-hydrogel interface. Based on the transient Stokes equation, we first demonstrate that the deviations are not due to finite particle radius, compressibility, or surface-slip effects. Small-angle neutron scattering rules out hydrogel heterogeneities. Instead, we show that a generalized Stokes-Einstein relation, accounting for an interfacial shell around tracers with viscoelastic properties that deviate from bulk, consistently explains our macrorheological and microrheological measurements. The extracted shell diameter is comparable to the PEO end-to-end distance, indicating the importance of dangling chain ends. Our methodology reveals the nanoscopic interfacial rheology of hydrogels and is applicable to different kinds of viscoelastic fluids and particles.

18.
Prog Polym Sci ; 1482024 Jan.
Article in English | MEDLINE | ID: mdl-38188703

ABSTRACT

The self-assembly of low-molecular-weight building motifs into supramolecular polymers has unlocked a new realm of materials with distinct properties and tremendous potential for advancing medical practices. Leveraging the reversible and dynamic nature of non-covalent interactions, these supramolecular polymers exhibit inherent responsiveness to their microenvironment, physiological cues, and biomolecular signals, making them uniquely suited for diverse biomedical applications. In this review, we intend to explore the principles of design, synthesis methodologies, and strategic developments that underlie the creation of supramolecular polymers as carriers for therapeutics, contributing to the treatment and prevention of a spectrum of human diseases. We delve into the principles underlying monomer design, emphasizing the pivotal role of non-covalent interactions, directionality, and reversibility. Moreover, we explore the intricate balance between thermodynamics and kinetics in supramolecular polymerization, illuminating strategies for achieving controlled sizes and distributions. Categorically, we examine their exciting biomedical applications: individual polymers as discrete carriers for therapeutics, delving into their interactions with cells, and in vivo dynamics; and supramolecular polymeric hydrogels as injectable depots, with a focus on their roles in cancer immunotherapy, sustained drug release, and regenerative medicine. As the field continues to burgeon, harnessing the unique attributes of therapeutic supramolecular polymers holds the promise of transformative impacts across the biomedical landscape.

19.
J Cell Physiol ; 239(2): e31165, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38149820

ABSTRACT

Transforming growth factor (TGF)-ß1 is a multifunctional cytokine that plays important roles in health and disease. Previous studies have revealed that TGFß1 activation, signaling, and downstream cell responses including epithelial-mesenchymal transition (EMT) and apoptosis are regulated by the elasticity or stiffness of the extracellular matrix. However, tissues within the body are not purely elastic, rather they are viscoelastic. How matrix viscoelasticity impacts cell fate decisions downstream of TGFß1 remains unknown. Here, we synthesized polyacrylamide hydrogels that mimic the viscoelastic properties of breast tumor tissue. We found that increasing matrix viscous dissipation reduces TGFß1-induced cell spreading, F-actin stress fiber formation, and EMT-associated gene expression changes, and promotes TGFß1-induced apoptosis in mammary epithelial cells. Furthermore, TGFß1-induced expression of integrin linked kinase (ILK) and colocalization of ILK with vinculin at cell adhesions is attenuated in mammary epithelial cells cultured on viscoelastic substrata in comparison to cells cultured on nearly elastic substrata. Overexpression of ILK promotes TGFß1-induced EMT and reduces apoptosis in cells cultured on viscoelastic substrata, suggesting that ILK plays an important role in regulating cell fate downstream of TGFß1 in response to matrix viscoelasticity.


Subject(s)
Extracellular Matrix , Signal Transduction , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Extracellular Matrix/metabolism , Integrins/metabolism , Transforming Growth Factor beta1/pharmacology , Transforming Growth Factor beta1/metabolism , Animals , Mice , Cell Line , Elasticity , Viscosity
20.
Adv Funct Mater ; 34(3)2024 Jan 15.
Article in English | MEDLINE | ID: mdl-39281808

ABSTRACT

Hydrogels have emerged as prototypical stimuli-responsive materials with potential applications in soft robotics, microfluidics, tissue engineering, and adaptive optics. To leverage the full potential of these materials, fabrication techniques capable of simultaneous control of microstructure, device architecture, and interfacial stability, i.e., adhesion of hydrogel components to support substrates, are needed. A universal strategy for the microfabrication of hydrogel-based devices with robust substrate adhesion amenable to use in liquid environments would enable numerous applications. This manuscript reports a general approach for the facile production of covalently attached, ordered arrays of microscale hydrogels (microgels) on silicone supports. Specifically, silicone-based templates were used to: i) drive mechanical assembly of prepolymer droplets into well-defined geometries and morphologies, and ii) present appropriate conjugation moieties to fix gels in place during photoinitiated crosslinking via a "graft from" polymerization scheme. Automated processing enabled rapid microgel array production for characterization, testing, and application. Furthermore, the stimuli-responsive microlensing properties of these arrays, via contractile modulated refractive index, were demonstrated. This process is directly applicable to the fabrication of adaptive optofluidic systems and can be further applied to advanced functional systems such as soft actuators and robotics and 3D cell culture technologies.

SELECTION OF CITATIONS
SEARCH DETAIL