Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.411
Filter
1.
J Nanobiotechnology ; 22(1): 378, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38943185

ABSTRACT

Tissue engineered heart valves (TEHVs) demonstrates the potential for tissue growth and remodel, offering particular benefit for pediatric patients. A significant challenge in designing functional TEHV lies in replicating the anisotropic mechanical properties of native valve leaflets. To establish a biomimetic TEHV model, we employed melt-electrowriting (MEW) technology to fabricate an anisotropic PCL scaffold. By integrating the anisotropic MEW-PCL scaffold with bioactive hydrogels (GelMA/ChsMA), we successfully crafted an elastic scaffold with tunable mechanical properties closely mirroring the structure and mechanical characteristics of natural heart valves. This scaffold not only supports the growth of valvular interstitial cells (VICs) within a 3D culture but also fosters the remodeling of extracellular matrix of VICs. The in vitro experiments demonstrated that the introduction of ChsMA improved the hemocompatibility and endothelialization of TEHV scaffold. The in vivo experiments revealed that, compared to their non-hydrogel counterparts, the PCL-GelMA/ChsMA scaffold, when implanted into SD rats, significantly suppressed immune reactions and calcification. In comparison with the PCL scaffold, the PCL-GelMA/ChsMA scaffold exhibited higher bioactivity and superior biocompatibility. The amalgamation of MEW technology and biomimetic design approaches provides a new paradigm for manufacturing scaffolds with highly controllable microstructures, biocompatibility, and anisotropic mechanical properties required for the fabrication of TEHVs.


Subject(s)
Heart Valves , Hydrogels , Rats, Sprague-Dawley , Tissue Engineering , Tissue Scaffolds , Tissue Engineering/methods , Animals , Tissue Scaffolds/chemistry , Anisotropy , Rats , Hydrogels/chemistry , Biocompatible Materials/chemistry , Heart Valve Prosthesis , Polyesters/chemistry , Cells, Cultured , Humans , Extracellular Matrix/chemistry , Male
2.
ACS Appl Mater Interfaces ; 16(25): 32078-32086, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38865735

ABSTRACT

The traditional recognition of extracellular matrix (ECM) at tissue sections relies on the time-consuming immunofluorescence that could not meet the demand of rapid diagnosis. Herein, we introduce a thickness-resolved electrochemiluminescence (ECL) microscopy to image thin-layer ECM at tissue sections for fast histopathological analysis. The unique surface-confined ECL mechanism enables to unveil the diversity and complexity of multiple tissue structures with varying thicknesses. Notably, the short lifetimes and the limited diffusion of electrogenerated coreactant radicals combined with their chemical reactivity result in a 2-fold increase in ECL intensity on ECM structures compared to the remaining tissue, enabling ECM visualization without specific labeling. The further quantitation of the ECM localization within tissue sections furnishes crucial insights into tumor progression and, more importantly, differentiates carcinoma and paracancerous tissues from patients in less than 30 min. Moreover, the reported electrochemistry-based microscopy is a dynamic approach allowing to investigate the transport, tortuosity, and trafficking properties through the tissues. This thickness-resolved recognition strategy not only opens new avenues for imaging complex samples but also holds promise for expediting tissue pathologic diagnosis, offering a more automated protocol with enhanced quantitative data compared to current intraoperative pathology methods.


Subject(s)
Electrochemical Techniques , Extracellular Matrix , Neoplasms , Humans , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Electrochemical Techniques/methods , Neoplasms/diagnosis , Neoplasms/pathology , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Luminescent Measurements/methods , Microscopy/methods
3.
Orthopadie (Heidelb) ; 53(7): 503-510, 2024 Jul.
Article in German | MEDLINE | ID: mdl-38829400

ABSTRACT

The various connective tissues of the body have different functions, which result from their specific structure and composition. The identification of this structure-function relationship is of great importance for various disciplines such as medicine, biology or tissue engineering. Connective tissue consists mainly of an extracellular matrix (ECM) and a limited number of cells. It is extremely adaptable because the activity of the cells remodels the composition and structure of the ECM in order to adapt the mechanical properties (functions) to the new demands (e.g. an increased mechanical stimulus).


Subject(s)
Connective Tissue , Extracellular Matrix , Connective Tissue/physiology , Humans , Extracellular Matrix/physiology , Extracellular Matrix/chemistry , Biomechanical Phenomena/physiology , Models, Biological , Animals , Tissue Engineering/methods
4.
J Am Chem Soc ; 146(26): 17539-17558, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38888174

ABSTRACT

Hydrogels have emerged as a promising class of extracellular matrix (ECM)-mimicking materials in regenerative medicine. Here, we briefly describe current state-of-the-art of ECM-mimicking hydrogels, ranging from natural to hybrid to completely synthetic versions, giving the prelude to the importance of supramolecular interactions to make true ECM mimics. The potential of supramolecular interactions to create ECM mimics for cell culture is illustrated through a focus on two different supramolecular hydrogel systems, both developed in our laboratories. We use some recent, significant findings to present important design principles underlying the cell-material interaction. To achieve cell spreading, we propose that slow molecular dynamics (monomer exchange within fibers) is crucial to ensure the robust incorporation of cell adhesion ligands within supramolecular fibers. Slow bulk dynamics (stress-relaxation─fiber rearrangements, τ1/2 ≈ 1000 s) is required to achieve cell spreading in soft gels (<1 kPa), while gel stiffness overrules dynamics in stiffer gels. Importantly, this resonates with the findings of others which specialize in different material types: cell spreading is impaired in case substrate relaxation occurs faster than clutch binding and focal adhesion lifetime. We conclude with discussing considerations and limitations of the supramolecular approach as well as provide a forward thinking perspective to further understand supramolecular hydrogel-cell interactions. Future work may utilize the presented guidelines underlying cell-material interactions to not only arrive at the next generation of ECM-mimicking hydrogels but also advance other fields, such as bioelectronics, opening up new opportunities for innovative applications.


Subject(s)
Extracellular Matrix , Hydrogels , Extracellular Matrix/chemistry , Hydrogels/chemistry , Humans , Cell Adhesion , Biomimetic Materials/chemistry
5.
Nano Lett ; 24(26): 7953-7961, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38888317

ABSTRACT

The physical properties of nanoscale cell-extracellular matrix (ECM) ligands profoundly impact biological processes, such as adhesion, motility, and differentiation. While the mechanoresponse of cells to static ligands is well-studied, the effect of dynamic ligand presentation with "adaptive" properties on cell mechanotransduction remains less understood. Utilizing a controllable diffusible ligand interface, we demonstrated that cells on surfaces with rapid ligand mobility could recruit ligands through activating integrin α5ß1, leading to faster focal adhesion growth and spreading at the early adhesion stage. By leveraging UV-light-sensitive anchor molecules to trigger a "dynamic to static" transformation of ligands, we sequentially activated α5ß1 and αvß3 integrins, significantly promoting osteogenic differentiation of mesenchymal stem cells. This study illustrates how manipulating molecular dynamics can directly influence stem cell fate, suggesting the potential of "sequentially" controlled mobile surfaces as adaptable platforms for engineering smart biomaterial coatings.


Subject(s)
Cell Adhesion , Cell Differentiation , Mechanotransduction, Cellular , Mesenchymal Stem Cells , Surface Properties , Mesenchymal Stem Cells/cytology , Humans , Integrin alpha5beta1/metabolism , Osteogenesis , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Integrin alphaVbeta3/metabolism , Ligands , Focal Adhesions
6.
ACS Appl Mater Interfaces ; 16(26): 32930-32944, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38888932

ABSTRACT

Protein materials are versatile tools in diverse biomedical fields. Among them, artificial secretory granules (SGs), mimicking those from the endocrine system, act as mechanically stable reservoirs for the sustained release of proteins as oligomeric functional nanoparticles. Only validated in oncology, the physicochemical properties of SGs, along with their combined drug-releasing and scaffolding abilities, make them suitable as smart topographies in regenerative medicine for the prolonged delivery of growth factors (GFs). Thus, considering the need for novel, safe, and cost-effective materials to present GFs, in this study, we aimed to biofabricate a protein platform combining both endocrine-like and extracellular matrix fibronectin-derived (ECM-FN) systems. This approach is based on the sustained delivery of a nanostructured histidine-tagged version of human fibroblast growth factor 2. The GF is presented onto polymeric surfaces, interacting with FN to spontaneously generate nanonetworks that absorb and present the GF in the solid state, to modulate mesenchymal stromal cell (MSC) behavior. The results show that SGs-based topographies trigger high rates of MSCs proliferation while preventing differentiation. While this could be useful in cell therapy manufacture demanding large numbers of unspecialized MSCs, it fully validates the hybrid platform as a convenient setup for the design of biologically active hybrid surfaces and in tissue engineering for the controlled manipulation of mammalian cell growth.


Subject(s)
Extracellular Matrix , Fibronectins , Mesenchymal Stem Cells , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Fibronectins/chemistry , Fibroblast Growth Factor 2/chemistry , Fibroblast Growth Factor 2/pharmacology , Cell Proliferation/drug effects , Cell Differentiation/drug effects , Nanostructures/chemistry
7.
Int J Biol Macromol ; 273(Pt 1): 132819, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38830498

ABSTRACT

The avascular nature of hyaline cartilage results in limited spontaneous self-repair and regenerative capabilities when damaged. Recent advances in three-dimensional bioprinting have enabled the precise dispensing of cell-laden biomaterials, commonly referred to as 'bioinks', which are emerging as promising solutions for tissue regeneration. An effective bioink for cartilage tissue engineering needs to create a micro-environment that promotes cell differentiation and supports neocartilage tissue formation. In this study, we introduced an innovative bioink composed of photocurable acrylated type I collagen (COLMA), thiol-modified hyaluronic acid (THA), and poly(ethylene glycol) diacrylate (PEGDA) for 3D bioprinting cartilage grafts using human nasal chondrocytes. Both collagen and hyaluronic acid, being key components of the extracellular matrix (ECM) in the human body, provide essential biological cues for tissue regeneration. We evaluated three formulations - COLMA, COLMA+THA, and COLMA+THA+PEGDA - for their printability, cell viability, structural integrity, and capabilities in forming cartilage-like ECM. The addition of THA and PEGDA significantly enhanced these properties, showcasing the potential of this bioink in advancing applications in cartilage repair and reconstructive surgery.


Subject(s)
Hyaluronic Acid , Tissue Engineering , Tissue Scaffolds , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Tissue Engineering/methods , Humans , Tissue Scaffolds/chemistry , Chondrocytes/cytology , Chondrocytes/drug effects , Polyethylene Glycols/chemistry , Bioprinting/methods , Collagen/chemistry , Printing, Three-Dimensional , Cartilage/cytology , Extracellular Matrix/chemistry , Cell Survival/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Ink
8.
Int J Biol Macromol ; 273(Pt 2): 133217, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38897519

ABSTRACT

Recent research focuses on fabricating scaffolds imitating the extracellular matrix (ECM) in texture, composition, and functionality. Moreover, specific nano-bio-particles can enhance cell differentiation. Decellularized ECM nanoparticles possess all of the mentioned properties. In this research, cartilage ECM, extracted from the cow's femur condyle, was decellularized, and ECM nanoparticles were synthesized. Finally, nanocomposite electrospun fibers containing polyhydroxybutyrate (PHB), chitosan (Cs) nanoparticles, and ECM nanoparticles were fabricated and characterized. TEM and DLS results revealed ECM nanoparticle sizes of 17.51 and 21.6 nm, respectively. Optimal performance was observed in the scaffold with 0.75 wt% ECM nanoparticles (PHB-Cs/0.75E). By adding 0.75 wt% ECM, the ultimate tensile strength and elongation at break increased by about 29 % and 21 %, respectively, while the water contact angle and crystallinity decreased by about 36° and 2 %, respectively. Uneven and rougher surfaces of the PHB-Cs/0.75E were determined by FESEM and AFM images, respectively. TEM images verified the uniform dispersion of nanoparticles within the fibers. After 70 days of degradation in PBS, the PHB-Cs/0.75E and PHB-Cs scaffolds demonstrated insignificant weight loss differences. Eventually, enhanced viability, attachment, and proliferation of the human costal chondrocytes on the PHB-Cs/0.75E scaffold, concluded from MTT, SEM, and DAPI staining, confirmed its potential for cartilage tissue engineering.


Subject(s)
Cartilage , Chitosan , Extracellular Matrix , Hydroxybutyrates , Nanoparticles , Tissue Engineering , Tissue Scaffolds , Chitosan/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Nanoparticles/chemistry , Animals , Hydroxybutyrates/chemistry , Cartilage/cytology , Cartilage/metabolism , Polyesters/chemistry , Humans , Cattle , Chondrocytes/cytology , Chondrocytes/metabolism , Polyhydroxybutyrates
9.
Food Res Int ; 189: 114549, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38876607

ABSTRACT

In cultured meat (CM) production, Scaffolding plays an important role by aiding cell adhesion, growth, differentiation, and alignment. The existence of fibrous microstructure in connective and muscle tissues has attracted considerable interest in the realm of tissue engineering and triggered the interest of researchers to implement scaffolding techniques. A wide array of research efforts is ongoing in scaffolding technologies for achieving the real meat structure on the principality of biomedical research and to replace serum free CM production. Scaffolds made of animal-derived biomaterials are found efficient in replicating the extracellular matrix (ECM), thus focus should be paid to utilize animal byproducts for this purpose. Proper identification and utilization of plant-derived scaffolding biomaterial could be helpful to add diversified options in addition to animal derived sources and reduce in cost of CM production through scaffolds. Furthermore, techniques like electrospinning, modified electrospinning and 3D bioprinting should be focused on to create 3D porous scaffolds to mimic the ECM of the muscle tissue and form real meat-like structures. This review discusses recent advances in cutting edge scaffolding techniques and edible biomaterials related to structured CM production.


Subject(s)
Extracellular Matrix , Tissue Engineering , Tissue Scaffolds , Tissue Scaffolds/chemistry , Tissue Engineering/methods , Animals , Extracellular Matrix/chemistry , Meat , Biocompatible Materials/chemistry , Bioprinting/methods , Printing, Three-Dimensional , In Vitro Meat
10.
ACS Appl Mater Interfaces ; 16(24): 30929-30957, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38832934

ABSTRACT

Bioengineered composite hydrogel platforms made of a supramolecular coassembly have recently garnered significant attention as promising biomaterial-based healthcare therapeutics. The mechanical durability of amyloids, in conjunction with the structured charged framework rendered by biologically abundant key ECM component glycosaminoglycan, enables us to design minimalistic customized biomaterial suited for stimuli responsive therapy. In this study, by harnessing the heparin sulfate-binding aptitude of amyloid fibrils, we have constructed a pH-responsive extracellular matrix (ECM) mimicking hydrogel matrix. This effective biocompatible platform comprising heparin sulfate-amyloid coassembled hydrogel embedded with polyphenol functionalized silver nanoparticles not only provide a native skin ECM-like conductive environment but also provide wound-microenvironment responsive on-demand superior antibacterial efficacy for effective diabetic wound healing. Interestingly, both the cytocompatibility and antibacterial properties of this bioinspired matrix can be fine-tuned by controlling the mutual ratio of heparin sulfate-amyloid and incubated silver nanoparticle components, respectively. The designed biomaterial platform exhibits notable effectiveness in the treatment of chronic hyperglycemic wounds infected with multidrug-resistant bacteria, because of the integration of pH-responsive release characteristics of the incubated functionalized AgNP and the antibacterial amyloid fibrils. In addition to this, the aforementioned assemblage shows exceptional hemocompatibility with significant antibiofilm and antioxidant characteristics. Histological evidence of the incised skin tissue sections indicates that the fabricated composite hydrogel is also effective in controlling pro-inflammatory cytokines such as IL6 and TNFα expressions at the wound vicinity with significant upregulation of angiogenesis markers like CD31 and α-SMA.


Subject(s)
Amyloid , Anti-Bacterial Agents , Extracellular Matrix , Heparin , Hydrogels , Metal Nanoparticles , Silver , Wound Healing , Wound Healing/drug effects , Hydrogels/chemistry , Hydrogels/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Heparin/chemistry , Heparin/pharmacology , Silver/chemistry , Silver/pharmacology , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Extracellular Matrix/drug effects , Metal Nanoparticles/chemistry , Amyloid/chemistry , Amyloid/metabolism , Animals , Humans , Staphylococcus aureus/drug effects , Escherichia coli/drug effects , Mice , Microbial Sensitivity Tests , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology
11.
Int J Biol Macromol ; 272(Pt 2): 132941, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38848842

ABSTRACT

Research in creating 3D structures mirroring the extracellular matrix (ECM) with accurate environmental cues holds paramount significance in biological applications.Biomaterials that replicate ECM properties-mechanical, physicochemical, and biological-emerge as pivotal tools in mimicking ECM behavior.Incorporating synthetic and natural biomaterials is widely used to produce scaffolds suitable for the intended organs.Polycaprolactone (PCL), a synthetic biomaterial, boasts commendable mechanical properties, albeit with relatively modest biological attributes due to its hydrophobic nature.Chitosan (CTS) exhibits strong biological traits but lacks mechanical resilience for complex tissue regeneration.Notably, both PCL and CTS have demonstrated their application in tissue engineering for diverse types of tissues.Their combination across varying PCL:CTS ratios has increased the likelihood of fabricating scaffolds to address defects in sturdy and pliable tissues.This comprehensive analysis aspires to accentuate their distinct attributes within tissue engineering across different organs.The central focus resides in the role of PCL:CTS-based scaffolds, elucidating their contribution to the evolution of advanced functional 3D frameworks tailored for tissue engineering across diverse organs.Moreover, this discourse delves into the considerations pertinent to each organ.


Subject(s)
Biocompatible Materials , Chitosan , Polyesters , Tissue Engineering , Tissue Scaffolds , Chitosan/chemistry , Tissue Engineering/methods , Polyesters/chemistry , Tissue Scaffolds/chemistry , Humans , Biocompatible Materials/chemistry , Animals , Extracellular Matrix/chemistry
12.
Biomed Mater ; 19(4)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38857607

ABSTRACT

Hypothyroidism is caused by insufficient stimulation or disruption of the thyroid. However, the drawbacks of thyroid transplantation have led to the search for new treatments. Decellularization allows tissue transplants to maintain their biomimetic structures while preserving cell adhesion, proliferation, and differentiation. This study aimed to decellularize human thyroid tissues using a structure-preserving optimization strategy and present preliminary data on recellularization. Nine methods were used for physical and chemical decellularization. Quantitative and immunohistochemical analyses were performed to investigate the DNA and extracellular matrix components of the tissues. Biomechanical properties were determined by compression test, and cell viability was examined after seeding MDA-T32 papillary thyroid cancer (PTC) cells onto the decellularized tissues. Decellularized tissues exhibited a notable decrease (<50 ng mg-1DNA, except for Groups 2 and 7) compared to the native thyroid tissue. Nonetheless, collagen and glycosaminoglycans were shown to be conserved in all decellularized tissues. Laminin and fibronectin were preserved at comparatively higher levels, and Young's modulus was elevated when decellularization included SDS. It was observed that the strain value in Group 1 (1.63 ± 0.14 MPa) was significantly greater than that in the decellularized tissues between Groups 2-9, ranging from 0.13 ± 0.03-0.72 ± 0.29 MPa. Finally, viability assessment demonstrated that PTC cells within the recellularized tissue groups successfully attached to the 3D scaffolds and sustained metabolic activity throughout the incubation period. We successfully established a decellularization optimization for human thyroid tissues, which has potential applications in tissue engineering and transplantation research. Our next goal is to conduct recellularization using the methods utilized in Group 1 and transplant the primary thyroid follicular cell-seeded tissues into anin vivoanimal model, particularly due to their remarkable 3D structural preservation and cell adhesion-promoting properties.


Subject(s)
Cell Survival , Extracellular Matrix , Thyroid Gland , Tissue Engineering , Tissue Scaffolds , Tissue Engineering/methods , Humans , Thyroid Gland/cytology , Extracellular Matrix/metabolism , Extracellular Matrix/chemistry , Tissue Scaffolds/chemistry , Collagen/chemistry , Cell Adhesion , Glycosaminoglycans/metabolism , Glycosaminoglycans/chemistry , Cell Line, Tumor , DNA , Elastic Modulus , Cell Proliferation , Thyroid Neoplasms/pathology , Decellularized Extracellular Matrix/chemistry , Laminin/chemistry , Biomechanical Phenomena , Cell Differentiation , Thyroid Cancer, Papillary/pathology , Fibronectins/chemistry , Fibronectins/metabolism
13.
Biotechnol J ; 19(6): e2300570, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38864387

ABSTRACT

This article primarily introduces a new treatment for liver fibrosis/cirrhosis. We developed a hepatic patch by combining decellularized liver matrix (DLM) with the hepatocyte growth factor (HGF)/heparin-complex and evaluated its restorative efficacy. In vitro prophylactic results, the HGF/heparin-DLM patches effectively mitigated CCl4-induced hepatocyte toxicity and restored the cytotoxicity levels to the baseline levels by day 5. Furthermore, these patches restored albumin synthesis of injured hepatocytes to more than 70% of the normal levels within 5 days. In vitro therapeutic results, the urea synthesis of the injured hepatocytes reached 91% of the normal levels after 10 days of culture, indicating successful restoration of hepatic function by the HGF/heparin-DLM patches in both prophylactic and therapeutic models. In vivo results, HGF/heparin-DLM patches attached to the liver and gut exhibited a significant decrease in collagen content (4.44 times and 2.77 times, respectively) and an increase in glycogen content (1.19 times and 1.12 times, respectively) compared to the fibrosis group after 1 week, separately. In summary, liver function was restored and inflammation was inhibited through the combined effects of DLM and the HGF/heparin-complex in fibrotic liver. The newly designed hepatic patch holds promise for both in vitro and in vivo regeneration therapy and preventive health care for liver tissue engineering.


Subject(s)
Carbon Tetrachloride , Heparin , Hepatocyte Growth Factor , Hepatocytes , Liver , Animals , Carbon Tetrachloride/toxicity , Hepatocyte Growth Factor/metabolism , Heparin/chemistry , Hepatocytes/drug effects , Male , Extracellular Matrix/metabolism , Extracellular Matrix/chemistry , Tissue Engineering/methods , Mice , Rats , Liver Cirrhosis/therapy , Chemical and Drug Induced Liver Injury/metabolism , Humans , Tissue Scaffolds/chemistry , Rats, Sprague-Dawley
14.
J Nanobiotechnology ; 22(1): 322, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38849858

ABSTRACT

The ideal tissue engineering scaffold should facilitate rapid cell infiltration and provide an optimal immune microenvironment during interactions with the host. Electrospinning can produce two-dimensional (2D) membranes mimicking the extracellular matrix. However, their dense structure hinders cell penetration, and their thin form restricts scaffold utility. In this study, latticed hydrogels were three-dimensional (3D) printed onto electrospun membranes. This technique allowed for layer-by-layer assembly of the membranes into 3D scaffolds, which maintained their resilience impressively under both dry and wet conditions. We assessed the cellular and host responses of these 3D nanofiber scaffolds by comparing random membranes and mesh-like membranes with three different mesh sizes (250, 500, and 750 µm). It was found that scaffolds with a mesh size of 500 µm were superior for M2 macrophage phenotype polarization, vascularization, and matrix deposition. Furthermore, it was confirmed by subsequent experiments such as RNA sequencing that the mesh-like topology may promote polarization to the M2 phenotype by affecting the PI3K/AKT pathway. In conclusion, our work offers a novel method for transforming 2D nanofiber membranes into 3D scaffolds. This method boasts flexibility, allowing for the use of varied electrospun membranes and hydrogels in terms of structure and composition. It has vast potential in tissue repair and regeneration.


Subject(s)
Hydrogels , Nanofibers , Printing, Three-Dimensional , Regenerative Medicine , Tissue Engineering , Tissue Scaffolds , Nanofibers/chemistry , Tissue Scaffolds/chemistry , Tissue Engineering/methods , Regenerative Medicine/methods , Hydrogels/chemistry , Animals , Mice , Macrophages/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/chemistry , RAW 264.7 Cells , Humans
15.
Anal Chim Acta ; 1315: 342798, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38879217

ABSTRACT

BACKGROUND: MMP-9 plays a crucial role in regulating the degradation of proteins within the extracellular matrix (ECM). This process closely correlates with the occurrence, development, invasion, and metastasis of various tumors, each exhibiting diverse levels of MMP-9 expression. However, the accuracy of detection results using the single-mode method is compromised due to the coexistence of multiple biologically active substances in the ECM. RESULTS: Therefore, in this study, a tri-modal detection system is proposed to obtain more accurate information by cross-verifying the results. Herein, we developed a tri-modal assay using the ZIF-8@Au NPs@S QDs composite as a multifunctional signal probe, decorated with DNA for the specific capture of MMP9. Notably, the probe demonstrated high conductivity, fluorescence response and mimicked enzyme catalytic activity. The capture segments of hybrid DNA specifically bind to MMP9 in the presence of MMP9, causing the signal probe to effortlessly detach the sensor interface onto the sample solution. Consequently, the sensor current performance is weakened, with the colorimetric and fluorescent signals becoming stronger with increasing MMP9 concentration. Notably, the detection range of the tri-modal sensor platform spans over 10 orders of magnitude, verifying notable observations of MMP-9 secretion in four tumor cell lines with chemotherapeutic drugs. Furthermore, the reliability of the detection results can be enhanced by employing pairwise comparative analysis. SIGNIFICANCE: This paper presents an effective strategy for detecting MMP9, which can be utilized for both the assessment of MMP-9 in cell lines and for analyzing the activity and mechanisms involved in various tumors.


Subject(s)
Antineoplastic Agents , Colorimetry , Electrochemical Techniques , Extracellular Matrix , Matrix Metalloproteinase 9 , Metal-Organic Frameworks , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/analysis , Humans , Colorimetry/methods , Electrochemical Techniques/methods , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Extracellular Matrix/metabolism , Extracellular Matrix/chemistry , Metal-Organic Frameworks/chemistry , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , Spectrometry, Fluorescence , Gold/chemistry , Biosensing Techniques/methods
16.
J Biosci ; 492024.
Article in English | MEDLINE | ID: mdl-38920105

ABSTRACT

Fibroblasts embedded in a 3D matrix microenvironment can remodel the matrix to regulate cell adhesion and function. Collagen hydrogels are a useful in vitro system to study cell-matrix interactions in a 3D microenvironment. While major matrix reorganizations are easily recognizable, subtle changes in response to environmental or biochemical cues are challenging to discern in 3D hydrogels. Three-dimensional collagen gels at 1.0 mg/ml vs 1.5 mg/ml were labelled with DQ-collagen and imaged by confocal reflectance microscopy to evaluate these small changes. An image analysis pipeline was developed, hydrogel area and number of crosssections analysed were optimized, and fibrillar collagen properties (number of branches, number of junctions, and average branch length) were quantified. While no significant changes were seen in fibrillar collagen organization between 1.0 mg/ml and 1.5 mg/ml collagen hydrogels, embedded mouse fibroblasts caused a significant increase in collagen branching and organization. Using the phalloidin-labelled cells, this change was quantitated in immediate proximity of the cell. A distinct increase in branch and junction numbers was observed, significantly altered by small changes in collagen concentration (1.0 mg/ml vs 1.5 mg/ml). Together, this analysis gives a quantitative evaluation of how cells respond to and modify their immediate microenvironment in a 3D collagen hydrogel.


Subject(s)
Fibroblasts , Hydrogels , Hydrogels/chemistry , Animals , Fibroblasts/metabolism , Fibroblasts/cytology , Mice , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Fibrillar Collagens/chemistry , Fibrillar Collagens/ultrastructure , Microscopy, Confocal , Collagen/chemistry , Cell Adhesion
17.
STAR Protoc ; 5(2): 103132, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38875112

ABSTRACT

The mandatory usage of extracellular matrix (ECM) gels in 3D cultures limits antibody penetration and increases background, while the removal of ECM gel causes disruption of morphology and sample loss. These factors pose challenges to effective immune labeling-based staining. Here, we present a protocol for whole-mount immunofluorescence staining of gel-embedded pancreatic organoids. We describe steps for sample fixation, blocking, and antibody incubation. We detail procedures for washing antibodies and mounting.


Subject(s)
Extracellular Matrix , Fluorescent Antibody Technique , Organoids , Pancreas , Organoids/cytology , Organoids/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/chemistry , Pancreas/cytology , Pancreas/metabolism , Fluorescent Antibody Technique/methods , Animals , Staining and Labeling/methods , Humans , Gels/chemistry , Mice
18.
Biomacromolecules ; 25(7): 4001-4013, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38814168

ABSTRACT

A major component of the extracellular matrix (ECM), laminins, modulates cells via diverse receptors. Their fragments have emerging utility as components of "ECM-mimetics" optimized to promote cell-based therapies. Recently, we reported that a bioactive laminin peptide known as A99 enhanced cell binding and spreading via fusion to an elastin-like polypeptide (ELP). The ELP "handle" serves as a rapid, noncovalent strategy to concentrate bioactive peptide mixtures onto a surface. We now report that this strategy can be further generalized across an expanded panel of additional laminin-derived elastin-like polypeptides (LELPs). A99 (AGTFALRGDNPQG), A2G80 (VQLRNGFPYFSY), AG73 (RKRLQVQLSIRT), and EF1m (LQLQEGRLHFMFD) all promote cell spreading while showing morphologically distinct F-actin formation. Equimolar mixtures of A99:A2G80-LELPs have synergistic effects on adhesion and spreading. Finally, three of these ECM-mimetics promote the neurite outgrowth of PC-12 cells. The evidence presented here demonstrates the potential of ELPs to deposit ECM-mimetics with applications in regenerative medicine, cell therapy, and tissue engineering.


Subject(s)
Cell Adhesion , Elastin , Laminin , Laminin/chemistry , Laminin/pharmacology , Elastin/chemistry , Animals , Rats , PC12 Cells , Cell Adhesion/drug effects , Extracellular Matrix/metabolism , Extracellular Matrix/chemistry , Peptides/chemistry , Peptides/pharmacology , Elastin-Like Polypeptides
19.
Biomater Adv ; 161: 213894, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38796956

ABSTRACT

Engineering of scaffolds for bone regeneration is often inspired by the native extracellular matrix mimicking its composite fibrous structure. In the present study, we used low loadings of diatomite earth (DE) biosilica to improve the bone regeneration potential of gelatin electrospun fibrillar microenvironments. We explored the effect of increasing the DE content from 1 % to 3 % and 5 %, respectively, on the physico-chemical properties of the fibrous scaffolds denoted FG_DE1, FG_DE3, FG_DE5, regarding the aqueous media affinity, stability under simulated physiological conditions, morphology characteristics, and local mechanical properties at the surface. The presence of biosilica generated composite structures with lower swelling degrees and higher stiffness when compared to gelatin fibers. Increasing DE content led to higher Young modulus, while the stability of the protein matrix in PBS, at 37 °C, over 21 was significantly decreased by the presence of diatomite loadings. The best preosteoblast response was obtained for FG_DE3, with enhanced mineralization during the osteogenic differentiation when compared to the control sample without diatomite. 5 % DE in FG_DE5 proved to negatively influence cells' metabolic activity and morphology. Hence, the obtained composite microfibrillar scaffolds might find application as osteoblast-responsive materials for bone tissue engineering.


Subject(s)
Gelatin , Osteoblasts , Tissue Engineering , Tissue Scaffolds , Gelatin/chemistry , Osteoblasts/drug effects , Osteoblasts/metabolism , Tissue Scaffolds/chemistry , Tissue Engineering/methods , Animals , Diatomaceous Earth/chemistry , Osteogenesis/drug effects , Cell Differentiation/drug effects , Mice , Bone Regeneration/drug effects , Cell Line , Cellular Microenvironment/drug effects , Microfibrils/chemistry , Microfibrils/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/chemistry , Extracellular Matrix/drug effects
20.
ACS Biomater Sci Eng ; 10(6): 3855-3867, 2024 06 10.
Article in English | MEDLINE | ID: mdl-38780042

ABSTRACT

Human corneal transplantation is still the only option to restore the function of corneal endothelial cells (CECs). Therefore, there is an urgent need for hCEC delivery systems to replace the human donor cornea. Here, we propose an alginate hydrogel (AH)-based delivery system, where a human fibroblast-derived, decellularized extracellular matrix (ECM) was physically integrated with AH. This AH securely combined with the ECM (ECM-AH) was approximately 50 µm thick, transparent, and permeable. The surface roughness and surface potential provided ECM-AH with a favorable microenvironment for CEC adhesion and growth in vitro. More importantly, ECM-AH could support the structural (ZO-1) and functional (Na+/K+-ATPase) markers of hCECs, as assessed via western blotting and quantitative polymerase chain reaction, which were comparable with those of a ferritic nitrocarburizing (FNC)-coated substrate (a positive control). The cell density per unit area was also significantly better with ECM-AH than the FNC substrate at day 7. A simulation test of cell engraftment in vitro showed that hCECs were successfully transferred into the decellularized porcine corneal tissue, where they were mostly alive. Furthermore, we found out that the endothelial-mesenchymal transition (EnMT)-inductive factors (Smad2 and vimentin) were largely declined with the hCECs grown on ECM-AH, whereas the EnMT inhibitory factor (Smad7) was significantly elevated. The difference was statistically significant compared to that of the FNC substrate. Moreover, we also observed that TGF-ß1-treated hCECs showed faster recovery of cell phenotype on the ECM. Taken together, our study demonstrates that ECM-AH is a very promising material for hCEC culture and delivery, which endows an excellent microenvironment for cell function and phenotype maintenance.


Subject(s)
Alginates , Extracellular Matrix , Fibroblasts , Hydrogels , Humans , Alginates/chemistry , Alginates/pharmacology , Hydrogels/chemistry , Hydrogels/pharmacology , Extracellular Matrix/metabolism , Extracellular Matrix/chemistry , Fibroblasts/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Endothelial Cells/metabolism , Endothelial Cells/cytology , Animals , Endothelium, Corneal/cytology , Endothelium, Corneal/metabolism , Epithelial-Mesenchymal Transition/drug effects , Swine , Cell Proliferation/drug effects , Endothelial-Mesenchymal Transition
SELECTION OF CITATIONS
SEARCH DETAIL
...