Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84.069
Filter
1.
Carbohydr Polym ; 337: 122144, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38710569

ABSTRACT

In vivo, cells interact with the extracellular matrix (ECM), which provides a multitude of biophysical and biochemical signals that modulate cellular behavior. Inspired by this, we explored a new methodology to develop a more physiomimetic polysaccharide-based matrix for 3D cell culture. Maleimide-modified alginate (AlgM) derivatives were successfully synthesized using DMTMM to activate carboxylic groups. Thiol-terminated cell-adhesion peptides were tethered to the hydrogel network to promote integrin binding. Rapid and efficient in situ hydrogel formation was promoted by thiol-Michael addition "click" chemistry via maleimide reaction with thiol-flanked protease-sensitive peptides. Alginate derivatives were further ionically crosslinked by divalent ions present in the medium, which led to greater stability and allowed longer cell culture periods. By tailoring alginate's biofunctionality we improved cell-cell and cell-matrix interactions, providing an ECM-like 3D microenvironment. We were able to systematically and independently vary biochemical and biophysical parameters to elicit specific cell responses, creating custom-made 3D matrices. DMTMM-mediated maleimide incorporation is a promising approach to synthesizing AlgM derivatives that can be leveraged to produce ECM-like matrices for a broad range of applications, from in vitro tissue modeling to tissue regeneration.


Subject(s)
Alginates , Click Chemistry , Extracellular Matrix , Hydrogels , Maleimides , Sulfhydryl Compounds , Maleimides/chemistry , Alginates/chemistry , Sulfhydryl Compounds/chemistry , Hydrogels/chemistry , Hydrogels/chemical synthesis , Extracellular Matrix/metabolism , Extracellular Matrix/chemistry , Humans , Cross-Linking Reagents/chemistry , Cell Adhesion/drug effects , Animals
2.
Food Res Int ; 186: 114396, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38729738

ABSTRACT

Cell culture meat is based on the scaled-up expansion of seed cells. The biological differences between seed cells from large yellow croakers in the two-dimensional (2D) and three-dimensional (3D) culture systems have not been explored. Here, satellite cells (SCs) from large yellow croakers (Larimichthys crocea) were grown on cell climbing slices, hydrogels, and microcarriers for five days to analyze the biological differences of SCs on different cell scaffolds. The results exhibited that SCs had different cell morphologies in 2D and 3D cultures. Cell adhesion receptors (Itgb1andsdc4) and adhesion spot markervclof the 3D cultures were markedly expressed. Furthermore, myogenic decision markers (Pax7andmyod) were significantly enhanced. However, the expression of myogenic differentiation marker (desmin) was significantly increased in the microcarrier group. Combined with the transcriptome data, this suggests that cell adhesion of SCs in 3D culture was related to the integrin signaling pathway. In contrast, the slight spontaneous differentiation of SCs on microcarriers was associated with rapid cell proliferation. This study is the first to report the biological differences between SCs in 2D and 3D cultures, providing new perspectives for the rapid expansion of cell culture meat-seeded cells and the development of customized scaffolds.


Subject(s)
Cell Adhesion , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Hydrogels , Satellite Cells, Skeletal Muscle , Tissue Scaffolds , Animals , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/cytology , Hydrogels/chemistry , Tissue Scaffolds/chemistry , Cell Culture Techniques, Three Dimensional/methods , Cells, Cultured , Desmin/metabolism , PAX7 Transcription Factor/metabolism , PAX7 Transcription Factor/genetics , Muscle Development
3.
Invest Ophthalmol Vis Sci ; 65(5): 4, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38691089

ABSTRACT

Purpose: To investigate the adhesion of Acanthamoeba to scleral contact lens (ScCL) surface according to lens shape. Methods: Two strains of A. polyphaga (CDC:V062 and ATCC 30461) and one clinical Acanthamoeba isolate, were inoculated onto five contact lens (CL): one first-generation silicone hydrogel (SHCL; lotrafilcon B; adhesion control) containing plasma surface treatment; two ScCL (fluorosilicone acrylate) one containing surface treatment composed of plasma and the other containing plasma with Hydra-PEG, and two CL designed with a flat shape having the same material and surface treatments of the ScCL. Trophozoites that adhered to the lens's surfaces were counted by inverted optical light microscopy. Possible alterations of the lens surface that could predispose amoeba adhesion and Acanthamoeba attached to these lens surfaces were evaluated by scanning electron microscopy (SEM). Results: All strains revealed greater adhesion to the ScCL when compared with the flat lenses (P < 0.001). The clinical isolate and the ATCC 30461 had a higher adhesion (P < 0.001) when compared with the CDC:V062. A rough texture was observed on the surface of the lenses that have been examined by SEM. Also, SEM revealed that the isolates had a rounded appearance on the surface of the ScCL in contrast with an elongated appearance on the surface of the silicone hydrogel. Conclusions: The findings revealed that the curved shape of the ScCL favors amoeba adhesion.


Subject(s)
Acanthamoeba , Microscopy, Electron, Scanning , Acanthamoeba/physiology , Acanthamoeba/ultrastructure , Sclera , Humans , Contact Lenses, Hydrophilic/parasitology , Cell Adhesion/physiology , Contact Lenses/parasitology , Trophozoites/ultrastructure , Trophozoites/physiology , Hydrogels , Animals
4.
J R Soc Interface ; 21(214): 20240022, 2024 May.
Article in English | MEDLINE | ID: mdl-38715321

ABSTRACT

Using a three-dimensional model of cell monolayers, we study the spatial organization of active stress chains as the monolayer transitions from a solid to a liquid state. The critical exponents that characterize this transition map the isotropic stress percolation onto the two-dimensional random percolation universality class, suggesting short-range stress correlations near this transition. This mapping is achieved via two distinct, independent pathways: (i) cell-cell adhesion and (ii) active traction forces. We unify our findings by linking the nature of this transition to high-stress fluctuations, distinctly linked to each pathway. The results elevate the importance of the transmission of mechanical information in dense active matter and provide a new context for understanding the non-equilibrium statistical physics of phase transition in active systems.


Subject(s)
Cell Adhesion , Models, Biological , Cell Adhesion/physiology , Stress, Mechanical , Phase Transition
5.
Sci Rep ; 14(1): 12212, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38806571

ABSTRACT

A positive family history is a major independent risk factor for atherosclerosis, and genetic variation is an important aspect of cardiovascular disease research. We identified a heterozygous missense variant p.L245P in the MMP10 gene in two families with premature myocardial infarction using whole-exome sequencing. The aim of this study was to investigate the consequences of this variant using in-silico and functional in-vitro assays. Molecular dynamics simulations were used to analyze protein interactions, calculate free binding energy, and measure the volume of the substrate-binding cleft of MMP10-TIMP1 models. The p.L245P variant showed an altered protein surface, different intra- and intermolecular interactions of MMP10-TIMP1, a lower total free binding energy between MMP10-TIMP1, and a volume-minimized substrate-binding cleft of MMP10 compared to the wild-type. For the functional assays, human THP-1 cells were transfected with plasmids containing MMP10 cDNA carrying the p.L245P and wild-type variant and differentiated into macrophages. Macrophage adhesion and migration assays were then conducted, and pro-inflammatory chemokine levels were evaluated. The p.L245P variant led to macrophages that were more adherent, less migratory, and secreted higher levels of the pro-inflammatory chemokines CXCL1 and CXCL8 than wild-type macrophages. Thus, the p.L245P variant in MMP10 may influence the pathogenesis of atherosclerosis in families with premature myocardial infarction by altering protein - protein interactions, macrophage adhesion and migration, and expression of pro-inflammatory chemokines, which may increase plaque rupture. These results could contribute to the development of selective MMP10 inhibitors and reduce the risk of atherosclerosis in families with a history of premature myocardial infarction.


Subject(s)
Matrix Metalloproteinase 10 , Mutation, Missense , Myocardial Infarction , Humans , Myocardial Infarction/genetics , Matrix Metalloproteinase 10/genetics , Matrix Metalloproteinase 10/metabolism , Male , Female , Pedigree , Adult , Molecular Dynamics Simulation , Macrophages/metabolism , THP-1 Cells , Middle Aged , Exome Sequencing , Cell Movement/genetics , Genetic Predisposition to Disease , Cell Adhesion/genetics , Tissue Inhibitor of Metalloproteinase-1
6.
Cells ; 13(10)2024 May 09.
Article in English | MEDLINE | ID: mdl-38786035

ABSTRACT

There are fewer investigations conducted on human primary endometrial epithelial cells (HPEECs) compared to human primary endometrial stromal cells (HPESCs). One of the main reasons is the scarcity of protocols enabling prolonged epithelial cell culture. Even though it is possible to culture HPEECs in 3D over a longer period of time, it is technically demanding. In this study, we successfully established a highly pure, stable, and long-term viable human conditionally reprogrammed endometrial epithelial cell line, designated as eCRC560. These cells stained positive for epithelial markers, estrogen and progesterone receptors, and epithelial cell-cell contacts but negative for stromal and endothelial cell markers. Estradiol (ES) reduced the abundance of ZO-1 in a time- and dose-dependent manner, in contrast to the dose-dependent increase with the progestin dienogest (DNG) when co-cultured with HPESCs. Moreover, ES significantly increased cell viability, cell migration, and invasion of the eCRC560 cells; all these effects were inhibited by pretreatment with DNG. DNG withdrawal led to a significantly disrupted monolayer of eCRC560 cells in co-culture with HPESCs, yet it markedly increased the adhesion of eCRC560 to the human mesothelial MeT-5A cells. The long-term viable eCRC560 cells are suitable for in vitro analysis of HPEECs to study the epithelial compartment of the human endometrium and endometrial pathologies.


Subject(s)
Cell Survival , Endometrium , Epithelial Cells , Estrogens , Progestins , Humans , Female , Endometrium/cytology , Endometrium/drug effects , Endometrium/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Progestins/pharmacology , Estrogens/pharmacology , Cell Survival/drug effects , Cell Movement/drug effects , Cell Line , Estradiol/pharmacology , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/cytology , Coculture Techniques , Time Factors , Cell Adhesion/drug effects
7.
J R Soc Interface ; 21(214): 20240105, 2024 May.
Article in English | MEDLINE | ID: mdl-38774959

ABSTRACT

During mesenchymal migration, F-actin protrusion at the leading edge and actomyosin contraction determine the retrograde flow of F-actin within the lamella. The coupling of this flow to integrin-based adhesions determines the force transmitted to the extracellular matrix and the net motion of the cell. In tissues, motion may also arise from convection, driven by gradients in tissue-scale surface tensions and pressures. However, how migration coordinates with convection to determine the net motion of cellular ensembles is unclear. To explore this, we study the spreading of cell aggregates on adhesive micropatterns on compliant substrates. During spreading, a cell monolayer expands from the aggregate towards the adhesive boundary. However, cells are unable to stabilize the protrusion beyond the adhesive boundary, resulting in retraction of the protrusion and detachment of cells from the matrix. Subsequently, the cells move upwards and rearwards, yielding a bulk convective flow towards the centre of the aggregate. The process is cyclic, yielding a steady-state balance between outward (protrusive) migration along the surface, and 'retrograde' (contractile) flows above the surface. Modelling the cell aggregates as confined active droplets, we demonstrate that the interplay between surface tension-driven flows within the aggregate, radially outward monolayer flow and conservation of mass leads to an internal circulation.


Subject(s)
Cell Adhesion , Cell Movement , Models, Biological , Cell Movement/physiology , Cell Adhesion/physiology , Cell Aggregation/physiology , Animals , Humans , Actins/metabolism
8.
Carbohydr Polym ; 338: 122204, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38763712

ABSTRACT

This study presents the development and characterization of a novel double-network self-healing hydrogel based on N-carboxyethyl chitosan (CEC) and oxidized dextran (OD) with the incorporation of crosslinked collagen (CEC-OD/COL-GP) to enhance its biological and physicochemical properties. The hydrogel formed via dynamic imine bond formation exhibited efficient self-healing within 30 min, and a compressive modulus recovery of 92 % within 2 h. In addition to its self-healing ability, CEC-OD/COL-GP possesses unique physicochemical characteristics including transparency, injectability, and adhesiveness to various substrates and tissues. Cell encapsulation studies confirmed the biocompatibility and suitability of the hydrogel as a cell-culture scaffold, with the presence of a collagen network that enhances cell adhesion, spreading, long-term cell viability, and proliferation. Leveraging their unique properties, we engineered assemblies of self-healing hydrogel modules for controlled spatiotemporal drug delivery and constructed co-culture models that simulate angiogenesis in tumor microenvironments. Overall, the CEC-OD/COL-GP hydrogel is a versatile and promising material for biomedical applications, offering a bottom-up approach for constructing complex structures with self-healing capabilities, controlled drug release, and support for diverse cell types in 3D environments. This hydrogel platform has considerable potential for advancements in tissue engineering and therapeutic interventions.


Subject(s)
Cell Adhesion , Chitosan , Dextrans , Hydrogels , Hydrogels/chemistry , Hydrogels/pharmacology , Chitosan/chemistry , Dextrans/chemistry , Humans , Cell Adhesion/drug effects , Cell Survival/drug effects , Collagen/chemistry , Animals , Drug Liberation , Cell Proliferation/drug effects , Cell Encapsulation/methods , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Mice , Biomimetics/methods , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Tissue Scaffolds/chemistry
9.
J Vis Exp ; (207)2024 May 03.
Article in English | MEDLINE | ID: mdl-38767378

ABSTRACT

Ultrashort self-assembling peptides (SAPs) can spontaneously form nanofibers that resemble the extracellular matrix. These fibers allow the formation of hydrogels that are biocompatible, biodegradable, and non-immunogenic. We have previously proven that SAPs, when biofunctionalized with protein-derived motifs, can mimic the extracellular matrix characteristics that support colorectal organoid formation. These biofunctional peptide hydrogels retain the original parent peptide's mechanical properties, tunability, and printability while incorporating cues that allow cell-matrix interactions to increase cell adhesion. This paper presents the protocols needed to evaluate and characterize the effects of various biofunctional peptide hydrogels on cell adhesion and lumen formation using an adenocarcinoma cancer cell line able to form colorectal cancer organoids cost-effectively. These protocols will help evaluate biofunctional peptide hydrogel effects on cell adhesion and luminal formation using immunostaining and fluorescence image analysis. The cell line used in this study has been previously utilized for generating organoids in animal-derived matrices.


Subject(s)
Colorectal Neoplasms , Hydrogels , Organoids , Peptides , Organoids/cytology , Humans , Colorectal Neoplasms/pathology , Cell Line, Tumor , Hydrogels/chemistry , Peptides/chemistry , Nanofibers/chemistry , Adenocarcinoma/pathology , Extracellular Matrix/chemistry , Cell Adhesion/physiology
10.
J Korean Med Sci ; 39(19): e156, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38769921

ABSTRACT

The process of cancer metastasis is dependent on the cancer cells' capacity to detach from the primary tumor, endure in a suspended state, and establish colonies in other locations. Anchorage dependence, which refers to the cells' reliance on attachment to the extracellular matrix (ECM), is a critical determinant of cellular shape, dynamics, behavior, and, ultimately, cell fate in nonmalignant and cancer cells. Anchorage-independent growth is a characteristic feature of cells resistant to anoikis, a programmed cell death process triggered by detachment from the ECM. This ability to grow and survive without attachment to a substrate is a crucial stage in the progression of metastasis. The recently discovered phenomenon named "adherent-to-suspension transition (AST)" alters the requirement for anchoring and enhances survival in a suspended state. AST is controlled by four transcription factors (IKAROS family zinc finger 1, nuclear factor erythroid 2, BTG anti-proliferation factor 2, and interferon regulatory factor 8) and can detach cells without undergoing the typical epithelial-mesenchymal transition. Notably, AST factors are highly expressed in circulating tumor cells compared to their attached counterparts, indicating their crucial role in the spread of cancer. Crucially, the suppression of AST substantially reduces metastasis while sparing primary tumors. These findings open up possibilities for developing targeted therapies that inhibit metastasis and emphasize the importance of AST, leading to a fundamental change in our comprehension of how cancer spreads.


Subject(s)
Neoplasm Metastasis , Neoplasms , Humans , Neoplasms/pathology , Cell Adhesion , Extracellular Matrix/metabolism , Epithelial-Mesenchymal Transition , Anoikis , Transcription Factors/metabolism
11.
Proc Natl Acad Sci U S A ; 121(22): e2314533121, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38776373

ABSTRACT

Nanoparticles tethered with vasculature-binding epitopes have been used to deliver the drug into injured or diseased tissues via the bloodstream. However, the extent that blood flow dynamics affects nanoparticle retention at the target site after adhesion needs to be better understood. This knowledge gap potentially underlies significantly different therapeutic efficacies between animal models and humans. An experimentally validated mathematical model that accurately simulates the effects of blood flow on nanoparticle adhesion and retention, thus circumventing the limitations of conventional trial-and-error-based drug design in animal models, is lacking. This paper addresses this technical bottleneck and presents an integrated mathematical method that derives heavily from a unique combination of a mechanics-based dispersion model for nanoparticle transport and diffusion in the boundary layers, an asperity model to account for surface roughness of endothelium, and an experimentally calibrated stochastic nanoparticle-cell adhesion model to describe nanoparticle adhesion and subsequent retention at the target site under external flow. PLGA-b-HA nanoparticles tethered with VHSPNKK peptides that specifically bind to vascular cell adhesion molecules on the inflamed vascular wall were investigated. The computational model revealed that larger particles perform better in adhesion and retention at the endothelium for the particle sizes suitable for drug delivery applications and within physiologically relevant shear rates. The computational model corresponded closely to the in vitro experiments which demonstrates the impact that model-based simulations can have on optimizing nanocarriers in vascular microenvironments, thereby substantially reducing in vivo experimentation as well as the development costs.


Subject(s)
Nanoparticles , Nanoparticles/chemistry , Humans , Ligands , Drug Delivery Systems/methods , Cell Adhesion , Animals , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry
12.
Cell Biochem Funct ; 42(4): e4058, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38783647

ABSTRACT

We aimed to evaluate the materials based on 4-methacryloxyethyl trimellitate anhydride/methyl methacrylate tri-n-butylborane (Super-bond [SB]) and nano hydroxyapatite (naHAp) for the repair of perforation at pulp chamber floor (PPF) in vitro and in vivo models. SB and naHAp were mixed in the mass ratio of 10% or 30% to produce naHAp/SB. Human periodontal ligament stem cells (HPDLSCs) were cultured on resin discs of SB or naHAp/SB to analyze the effects of naHAp/SB on cell adhesion, proliferation, and cementoblastic differentiation. A rat PPF model was treated with SB or naHAp/SB to examine the effects of naHAp/SB on the healing of defected cementum and periodontal ligament (PDL) at the site of PPF. HPDLSCs were spindle-shaped and adhered to all resin discs. Changing the resin from SB to naHAp/SB did not significantly alter cell proliferation. Both 10% and 30% naHAp/SB were more effective than SB in promoting cementoblastic differentiation of HPDLSCs. In the rat PPF model, 30% naHAp/SB was more effective than SB in promoting the formation Sharpey's fiber-like structures with expression of the PDL-related marker and cementum-like structures with expression of cementum-related markers. In conclusion, 30% naHAp/SB can be the new restorative material for PPF because it exhibited the abilities of adhering to dentin and healing of defected periodontal tissue.


Subject(s)
Boron Compounds , Durapatite , Methacrylates , Periodontal Ligament , Animals , Rats , Humans , Durapatite/chemistry , Durapatite/pharmacology , Periodontal Ligament/drug effects , Periodontal Ligament/cytology , Periodontal Ligament/metabolism , Boron Compounds/pharmacology , Boron Compounds/chemistry , Methacrylates/chemistry , Methacrylates/pharmacology , Cell Differentiation/drug effects , Wound Healing/drug effects , Male , Cell Proliferation/drug effects , Dental Pulp Cavity/metabolism , Dental Pulp Cavity/drug effects , Stem Cells/drug effects , Stem Cells/cytology , Stem Cells/metabolism , Cells, Cultured , Rats, Sprague-Dawley , Methylmethacrylates/chemistry , Methylmethacrylates/pharmacology , Cell Adhesion/drug effects
13.
Commun Biol ; 7(1): 617, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38778159

ABSTRACT

The question of whether material stiffness enhances cell adhesion and clustering is still open to debate. Results from the literature are seemingly contradictory, with some reports illustrating that adhesion increases with surface stiffness and others suggesting that the performance of a system of cells is curbed by high values of elasticity. To address the role of elasticity as a regulator in neuronal cell adhesion and clustering, we investigated the topological characteristics of networks of neurons on polydimethylsiloxane (PDMS) surfaces - with values of elasticity (E) varying in the 0.55-2.65 MPa range. Results illustrate that, as elasticity increases, the number of neurons adhering on the surface decreases. Notably, the small-world coefficient - a topological measure of networks - also decreases. Numerical simulations and functional multi-calcium imaging experiments further indicated that the activity of neuronal cells on soft surfaces improves for decreasing E. Experimental findings are supported by a mathematical model, that explains adhesion and clustering of cells on soft materials as a function of few parameters - including the Young's modulus and roughness of the material. Overall, results indicate that - in the considered elasticity interval - increasing the compliance of a material improves adhesion, improves clustering, and enhances communication of neurons.


Subject(s)
Cell Adhesion , Elasticity , Neurons , Neurons/physiology , Animals , Dimethylpolysiloxanes/chemistry , Surface Properties , Elastic Modulus , Cells, Cultured , Rats
14.
Sci Rep ; 14(1): 11719, 2024 05 22.
Article in English | MEDLINE | ID: mdl-38778185

ABSTRACT

Cell adhesion experiments are important in tissue engineering and for testing new biologically active surfaces, prostheses, and medical devices. Additionally, the initial state of adhesion (referred to as nascent adhesion) plays a key role and is currently being intensively researched. A critical step in handling all adherent cell types is their dissociation from their substrates for further processing. Various cell dissociation methods and reagents are used in most tissue culture laboratories (here, cell dissociation from the culture surface, cell harvesting, and cell detachment are used interchangeably). Typically, the dissociated cells are re-adhered for specific measurements or applications. However, the impact of the choice of dissociation method on cell adhesion in subsequent measurements, especially when comparing the adhesivity of various surfaces, is not well clarified. In this study, we demonstrate that the application of a label-free optical sensor can precisely quantify the effect of cell dissociation methods on cell adhesivity, both at the single-cell and population levels. The optical measurements allow for high-resolution monitoring of cellular adhesion without interfering with the physiological state of the cells. We found that the choice of reagent significantly alters cell adhesion on various surfaces. Our results clearly demonstrate that biological conclusions about cellular adhesion when comparing various surfaces are highly dependent on the employed dissociation method. Neglecting the choice of cellular dissociation can lead to misleading conclusions when evaluating cell adhesion data from various sources and comparing the adhesivity of two different surfaces (i.e., determining which surface is more or less adhesive).


Subject(s)
Cell Adhesion , Humans , Surface Properties
15.
Yakugaku Zasshi ; 144(5): 497-501, 2024.
Article in Japanese | MEDLINE | ID: mdl-38692923

ABSTRACT

Signal-transducing adaptor protein-2 (STAP-2) is a unique scaffold protein that regulates several immunological signaling pathways, including LIF/LIF receptor and LPS/TLR4 signals. STAP-2 is required for Fas/FasL-dependent T cell apoptosis and SDF-1α-induced T cell migration. Conversely, STAP-2 modulates integrin-mediated T cell adhesion, suggesting that STAP-2 is essential for several negative and positive T cell functions. However, whether STAP-2 is involved in T cell-antigen receptor (TCR)-mediated T cell activation is unknown. STAP-2 deficiency was recently reported to suppress TCR-mediated T cell activation by inhibiting LCK-mediated CD3ζ and ZAP-70 activation. Using STAP-2 deficient mice, it was demonstrated that STAP-2 is required for the pathogenesis of Propionibacterium acnes-induced granuloma formation and experimental autoimmune encephalomyelitis. Here, detailed functions of STAP-2 in TCR-mediated T cell activation, and how STAP-2 affects the pathogenesis of T cell-mediated inflammation and immune diseases, are reviewed.


Subject(s)
Adaptor Proteins, Signal Transducing , Lymphocyte Activation , Receptors, Antigen, T-Cell , Signal Transduction , T-Lymphocytes , ZAP-70 Protein-Tyrosine Kinase , Animals , Receptors, Antigen, T-Cell/physiology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Humans , Adaptor Proteins, Signal Transducing/physiology , Adaptor Proteins, Signal Transducing/metabolism , Mice , ZAP-70 Protein-Tyrosine Kinase/metabolism , ZAP-70 Protein-Tyrosine Kinase/physiology , Propionibacterium acnes/physiology , Propionibacterium acnes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/etiology , Inflammation/immunology , Apoptosis , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Cell Movement , Cell Adhesion , CD3 Complex , Chemokine CXCL12/physiology , Chemokine CXCL12/metabolism
16.
Phys Rev Lett ; 132(18): 188402, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38759206

ABSTRACT

Cell adhesion receptors are transmembrane proteins that bind cells to their environment. These proteins typically cluster into disk-shaped or linear structures. Here, we show that such clustering patterns spontaneously emerge when the receptor senses the membrane deformation gradient, for example, by reaching a lower-energy conformation when the membrane is tilted relative to the underlying binding substrate. Increasing the strength of the membrane gradient-sensing mechanism first yields isolated disk-shaped clusters and then long linear structures. Our theory is coherent with experimental estimates of the parameters, suggesting that a tilt-induced clustering mechanism is relevant in the context of cell adhesion.


Subject(s)
Cell Membrane , Cell Membrane/metabolism , Models, Biological , Cell Adhesion/physiology , Phase Separation , Platelet Glycoprotein GPIb-IX Complex
17.
Nat Commun ; 15(1): 4160, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755128

ABSTRACT

The regeneration of critical-size bone defects, especially those with irregular shapes, remains a clinical challenge. Various biomaterials have been developed to enhance bone regeneration, but the limitations on the shape-adaptive capacity, the complexity of clinical operation, and the unsatisfied osteogenic bioactivity have greatly restricted their clinical application. In this work, we construct a mechanically robust, tailorable and water-responsive shape-memory silk fibroin/magnesium (SF/MgO) composite scaffold, which is able to quickly match irregular defects by simple trimming, thus leading to good interface integration. We demonstrate that the SF/MgO scaffold exhibits excellent mechanical stability and structure retention during the degradative process with the potential for supporting ability in defective areas. This scaffold further promotes the proliferation, adhesion and migration of osteoblasts and the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) in vitro. With suitable MgO content, the scaffold exhibits good histocompatibility, low foreign-body reactions (FBRs), significant ectopic mineralisation and angiogenesis. Skull defect experiments on male rats demonstrate that the cell-free SF/MgO scaffold markedly enhances bone regeneration of cranial defects. Taken together, the mechanically robust, personalised and bioactive scaffold with water-responsive shape-memory may be a promising biomaterial for clinical-size and irregular bone defect regeneration.


Subject(s)
Biocompatible Materials , Bone Regeneration , Fibroins , Magnesium , Mesenchymal Stem Cells , Osteogenesis , Tissue Scaffolds , Fibroins/chemistry , Fibroins/pharmacology , Bone Regeneration/drug effects , Animals , Tissue Scaffolds/chemistry , Male , Osteogenesis/drug effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Rats , Magnesium/chemistry , Magnesium/pharmacology , Biocompatible Materials/chemistry , Osteoblasts/drug effects , Cell Differentiation/drug effects , Rats, Sprague-Dawley , Water/chemistry , Cell Proliferation/drug effects , Tissue Engineering/methods , Skull/drug effects , Cell Adhesion/drug effects , Bombyx
18.
Sci Rep ; 14(1): 11231, 2024 05 16.
Article in English | MEDLINE | ID: mdl-38755203

ABSTRACT

Selecting and isolating various cell types is a critical procedure in many applications, including immune therapy, regenerative medicine, and cancer research. Usually, these selection processes involve some labeling or another invasive step potentially affecting cellular functionality or damaging the cell. In the current proof of principle study, we first introduce an optical biosensor-based method capable of classification between healthy and numerous cancerous cell types in a label-free setup. We present high classification accuracy based on the monitored single-cell adhesion kinetic signals. We developed a high-throughput data processing pipeline to build a benchmark database of ~ 4500 single-cell adhesion measurements of a normal preosteoblast (MC3T3-E1) and various cancer (HeLa, LCLC-103H, MDA-MB-231, MCF-7) cell types. Several datasets were used with different cell-type selections to test the performance of deep learning-based classification models, reaching above 70-80% depending on the classification task. Beyond testing these models, we aimed to draw interpretable biological insights from their results; thus, we applied a deep neural network visualization method (grad-CAM) to reveal the basis on which these complex models made their decisions. Our proof-of-concept work demonstrated the success of a deep neural network using merely label-free adhesion kinetic data to classify single mammalian cells into different cell types. We propose our method for label-free single-cell profiling and in vitro cancer research involving adhesion. The employed label-free measurement is noninvasive and does not affect cellular functionality. Therefore, it could also be adapted for applications where the selected cells need further processing, such as immune therapy and regenerative medicine.


Subject(s)
Cell Adhesion , Single-Cell Analysis , Humans , Single-Cell Analysis/methods , Kinetics , Mice , Animals , Biosensing Techniques/methods , Cell Line, Tumor
19.
ACS Nano ; 18(19): 12341-12354, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38695772

ABSTRACT

The patch with a superlubricated surface shows great potential for the prevention of postoperative adhesion during soft tissue repair. However, the existing patches suffer from the destruction of topography during superlubrication coating and lack of pro-healing capability. Herein, we demonstrate a facile and versatile strategy to develop a Janus nanofibrous patch (J-NFP) with antiadhesion and reactive oxygen species (ROS) scavenging functions. Specifically, sequential electrospinning is performed with initiators and CeO2 nanoparticles (CeNPs) embedded on the different sides, followed by subsurface-initiated atom transfer radical polymerization for grafting zwitterionic polymer brushes, introducing superlubricated skin on the surface of single nanofibers. The poly(sulfobetaine methacrylate) brush-grafted patch retains fibrous topography and shows a coefficient of friction of around 0.12, which is reduced by 77% compared with the pristine fibrous patch. Additionally, a significant reduction in protein, platelet, bacteria, and cell adhesion is observed. More importantly, the CeNPs-embedded patch enables ROS scavenging as well as inhibits pro-inflammatory cytokine secretion and promotes anti-inflammatory cytokine levels. Furthermore, the J-NFP can inhibit tissue adhesion and promote repair of both rat skin wounds and intrauterine injuries. The present strategy for developing the Janus patch exhibits enormous prospects for facilitating soft tissue repair.


Subject(s)
Nanofibers , Animals , Rats , Nanofibers/chemistry , Wound Healing/drug effects , Reactive Oxygen Species/metabolism , Skin/drug effects , Skin/pathology , Tissue Adhesions/prevention & control , Rats, Sprague-Dawley , Cell Adhesion/drug effects , Cerium/chemistry , Cerium/pharmacology , Surface Properties , Mice , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology
20.
Molecules ; 29(9)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38731542

ABSTRACT

Bilayer electrospun fibers aimed to be used for skin tissue engineering applications were fabricated for enhanced cell attachment and proliferation. Different ratios of PHBV-PLLA (70:30, 80:20, and 90:10 w/w) blends were electrospun on previously formed electrospun PHBV membranes to produce their bilayers. The fabricated electrospun membranes were characterized with FTIR, which conformed to the characteristic peaks assigned for both PHBV and PLLA. The surface morphology was evaluated using SEM analysis that showed random fibers with porous morphology. The fiber diameter and pore size were measured in the range of 0.7 ± 0.1 µm and 1.9 ± 0.2 µm, respectively. The tensile properties of the bilayers were determined using an electrodynamic testing system. Bilayers had higher elongation at break (44.45%) compared to the monolayers (28.41%) and improved ultimate tensile strength (7.940 MPa) compared to the PHBV monolayer (2.450 MPa). In vitro cytotoxicity of each of the scaffolds was determined via culturing MC3T3 (pre-osteoblastic cell line) on the membranes. Proliferation was evaluated using the Alamar Blue assay on days 3, 7, and 14, respectively. SEM images of cells cultured on membranes were taken in addition to bright field imaging to visually show cell attachment. Fluorescent nuclear staining performed with DAPI was imaged with an inverted fluorescent microscope. The fabricated bilayer shows high mechanical strength as well as biocompatibility with good cell proliferation and cell attachment, showing potential for skin substitute applications.


Subject(s)
Biocompatible Materials , Cell Proliferation , Polyesters , Skin , Tissue Engineering , Tissue Scaffolds , Tissue Engineering/methods , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Polyesters/chemistry , Animals , Mice , Cell Proliferation/drug effects , Tissue Scaffolds/chemistry , Tensile Strength , Membranes, Artificial , Cell Line , Materials Testing , Polymers/chemistry , Cell Adhesion/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...