Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 703
Filter
2.
Biomimetics (Basel) ; 9(7)2024 Jul 10.
Article in English | MEDLINE | ID: mdl-39056864

ABSTRACT

Ti6Al4V superalloy is recognized as a good candidate for bone implants owing to its biocompatibility, corrosion resistance, and high strength-to-weight ratio. While dense metal implants are associated with stress shielding issues due to the difference in densities, stiffness, and modulus of elasticity compared to bone tissues, the surface of the implant/scaffold should mimic the properties of the bone of interest to assure a good integration with a strong interface. In this study, we investigated the additive manufacturing of porous Ti6Al4V scaffolds and coating modification for enhanced osteoconduction using osteoblast cells. The results showed the successful fabrication of porous Ti6Al4V scaffolds with adequate strength. Additionally, the surface treatment with NaOH and Dopamine Hydrochloride (DOPA) promoted the formation of Dopamine Hydrochloride (DOPA) coating with an optimized coating process, providing an environment that supports higher cell viability and growth compared to the uncoated Ti6Al4V scaffolds, as demonstrated by the higher proliferation ratios observed from day 1 to day 29. These findings bring valuable insights into the surface modification of 3D-printed scaffolds for improved osteoconduction through the coating process in solutions.

3.
Appl Mater Today ; 382024 Jun.
Article in English | MEDLINE | ID: mdl-39006868

ABSTRACT

The conventional technique for successful bone grafts, involving the use of a patients own tissue (autografts), is challenged by limited availability and donor site morbidity. While allografts and xenografts offer alternatives, they come with the risk of rejection. This underscores the pressing need for tailor-made artificial bone graft materials. In this context, injectable hydrogels are emerging as a promising solution for bone regeneration, especially in complex maxillofacial reconstruction cases. These hydrogels can seamlessly adapt to irregular shapes and conservatively fill defects. Our study introduces a shear-thinning biomaterial by blending silicate nanoplatelets (SNs) enriched with human blood-derived plasma rich in growth factors (PRGF) for personalized applications. Notably, our investigations unveil that injectable hydrogel formulations comprising 7.5% PRGF yield sustained protein and growth factor release, affording precise control over critical growth factors essential for tissue regeneration. Moreover, our hydrogel exhibits exceptional biocompatibility in vitro and in vivo and demonstrates hemostatic properties. The hydrogel also presents a robust angiogenic potential and an inherent capacity to promote bone differentiation, proven through Alizarin Red staining, gene expression, and immunostaining assessments of bone-related biomarkers. Given these impressive attributes, our hydrogel stands out as a leading candidate for maxillofacial bone regeneration application. Beyond this, our findings hold immense potential in revolutionizing the field of regenerative medicine, offering an influential platform for crafting precise and effective therapeutic strategies.

4.
Article in English | MEDLINE | ID: mdl-39046205

ABSTRACT

In addressing the intricate challenges of enterocutaneous fistula (ECF) treatment, such as internal bleeding, effluent leakage, inflammation, and infection, our research is dedicated to introducing a regenerative adhesive hydrogel that can seal and expedite the healing process. A double syringe setup was utilized, with dopagelatin and platelet-rich plasma (PRP) in one syringe and Laponite and sodium periodate in another. The hydrogel begins to cross-link immediately after passing through a mixing tip and exhibits tissue adhesive properties. Results demonstrated that PRP deposits within the pores of the cross-linked hydrogel and releases sustainably, enhancing its regenerative capabilities. The addition of PRP further improved the mechanical properties and slowed down the degradation of the hydrogel. Furthermore, the hydrogel demonstrated cytocompatibility, hemostatic properties, and time-dependent macrophage M1 to M2 phase transition, suggesting the anti-inflammatory response of the material. In an in vitro bench test simulating high-pressure fistula conditions, the hydrogel effectively occluded pressures up to 300 mmHg. In conclusion, this innovative hydrogel holds promise for ECF treatment and diverse fistula cases, marking a significant advancement in its therapeutic approaches.

5.
Nat Commun ; 15(1): 5891, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003263

ABSTRACT

Synthetic Notch (synNotch) receptors are genetically encoded, modular synthetic receptors that enable mammalian cells to detect environmental signals and respond by activating user-prescribed transcriptional programs. Although some materials have been modified to present synNotch ligands with coarse spatial control, applications in tissue engineering generally require extracellular matrix (ECM)-derived scaffolds and/or finer spatial positioning of multiple ligands. Thus, we develop here a suite of materials that activate synNotch receptors for generalizable engineering of material-to-cell signaling. We genetically and chemically fuse functional synNotch ligands to ECM proteins and ECM-derived materials. We also generate tissues with microscale precision over four distinct reporter phenotypes by culturing cells with two orthogonal synNotch programs on surfaces microcontact-printed with two synNotch ligands. Finally, we showcase applications in tissue engineering by co-transdifferentiating fibroblasts into skeletal muscle or endothelial cell precursors in user-defined micropatterns. These technologies provide avenues for spatially controlling cellular phenotypes in mammalian tissues.


Subject(s)
Cell Differentiation , Receptors, Notch , Signal Transduction , Tissue Engineering , Receptors, Notch/metabolism , Tissue Engineering/methods , Animals , Humans , Mice , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Fibroblasts/cytology , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/genetics , Ligands , Tissue Scaffolds/chemistry , Muscle, Skeletal/metabolism , Muscle, Skeletal/cytology , Endothelial Cells/metabolism , Endothelial Cells/cytology , HEK293 Cells
6.
Aggregate (Hoboken) ; 5(2)2024 Apr.
Article in English | MEDLINE | ID: mdl-38800607

ABSTRACT

mRNA therapy is the intracellular delivery of messenger RNA (mRNA) to produce desired therapeutic proteins. Developing strategies for local mRNA delivery is still required where direct intra-articular injections are inappropriate for targeting a specific tissue. The mRNA delivery efficiency depends on protecting nucleic acids against nuclease-mediated degradation and safe site-specific intracellular delivery. Herein, we report novel mRNA-releasing matrices based on RGD-moiety-rich gelatin methacryloyl (GelMA) microporous annealed particle (MAP) scaffolds. GelMA concentration in aerogel-based microgels (µgels) produced through a microfluidic process, MAP stiffnesses, and microporosity are crucial parameters for cell adhesion, spreading, and proliferation. After being loaded with mRNA complexes, MAP scaffolds composed of 10 % GelMA µgels display excellent cell viability with increasing cell infiltration, adhesion, proliferation, and gene transfer. The intracellular delivery is achieved by the sustained release of mRNA complexes from MAP scaffolds and cell adhesion on mRNA-releasing scaffolds. These findings highlight that hybrid systems can achieve efficient protein expression by delivering mRNA complexes, making them promising mRNA-releasing biomaterials for tissue engineering.

7.
Adv Healthc Mater ; : e2400881, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38781005

ABSTRACT

Remote health monitoring and treatment serve as critical drivers for advancing health equity, bridging geographical and socioeconomic disparities, ensuring equitable access to quality healthcare for those in underserved or remote regions. By democratizing healthcare, this approach offers timely interventions, continuous monitoring, and personalized care independent of one's location or socioeconomic status, thereby striving for an equitable distribution of health resources and outcomes. Meanwhile, microneedle arrays (MNAs), revolutionize painless and minimally invasive access to interstitial fluid for drug delivery and diagnostics. This paper introduces an integrated theranostic MNA system employing an array of colorimetric sensors to quantitatively measure -pH, glucose, and lactate, alongside a remotely-triggered system enabling on-demand drug delivery. Integration of an ultrasonic atomizer streamlines the drug delivery, facilitating rapid, pumpless, and point-of-care drug delivery, enhancing system portability while reducing complexities. An accompanying smartphone application interfaces the sensing and drug delivery components. Demonstrated capabilities include detecting pH (3 to 8), glucose (up to 16 mm), and lactate (up to 1.6 mm), showcasing on-demand drug delivery, and assessing delivery system performance via a scratch assay. This innovative approach confronts drug delivery challenges, particularly in managing chronic diseases requiring long-term treatment, while also offering avenues for non-invasive health monitoring through microneedle-based sensors.

8.
Acta Biomater ; 183: 89-100, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38801867

ABSTRACT

Self-assembling peptide-based hydrogels have become a highly attractive scaffold for three-dimensional (3D) in vitro disease modeling as they provide a way to create tunable matrices that can resemble the extracellular matrix (ECM) of various microenvironments. Alzheimer's disease (AD) is an exceptionally complex neurodegenerative condition; however, our understanding has advanced due to the transition from two-dimensional (2D) to 3D in vitro modeling. Nonetheless, there is a current gap in knowledge regarding the role of amyloid structures, and previously developed models found long-term difficulty in creating an appropriate model involving the ECM and amyloid aggregates. In this report, we propose a multi-component self-assembling peptide-based hydrogel scaffold to mimic the amyloid-beta (ß) containing microenvironment. Characterization of the amyloid-ß-mimicking hydrogel (Col-HAMA-FF) reveals the formation of ß-sheet structures as a result of the self-assembling properties of phenylalanine (Phe, F) through π-π stacking of the residues, thus mimicking the amyloid-ß protein nanostructures. We investigated the effect of the amyloid-ß-mimicking microenvironment on healthy neuronal progenitor cells (NPCs) compared to a natural-mimicking matrix (Col-HAMA). Our results demonstrated higher levels of neuroinflammation and apoptosis markers when NPCs were cultured in the amyloid-like matrix compared to a natural brain matrix. Here, we provided insights into the impact of amyloid-like structures on NPC phenotypes and behaviors. This foundational work, before progressing to more complex plaque models, provides a promising scaffold for future investigations on AD mechanisms and drug testing. STATEMENT OF SIGNIFICANCE: In this study, we engineered two multi-component hydrogels: one to mimic the natural extracellular matrix (ECM) of the brain and one to resemble an amyloid-like microenvironment using a self-assembling peptide hydrogel. The self-assembling peptide mimics ß-amyloid fibrils seen in amyloid-ß protein aggregates. We report on the culture of neuronal progenitor cells within the amyloid-mimicking ECM scaffold to study the impact through marker expressions related to inflammation and DNA damage. This foundational work, before progressing to more complex plaque models, offers a promising scaffold for future investigations on AD mechanisms and drug testing.


Subject(s)
Amyloid beta-Peptides , Hydrogels , Neural Stem Cells , Hydrogels/chemistry , Hydrogels/pharmacology , Amyloid beta-Peptides/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Phenotype , Humans , Extracellular Matrix/metabolism , Extracellular Matrix/chemistry , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Animals
9.
ACS Sens ; 9(5): 2334-2345, 2024 05 24.
Article in English | MEDLINE | ID: mdl-38639453

ABSTRACT

Noninvasive monitoring of biofabricated tissues during the biomanufacturing process is needed to obtain reproducible, healthy, and functional tissues. Measuring the levels of biomarkers secreted from tissues is a promising strategy to understand the status of tissues during biofabrication. Continuous and real-time information from cultivated tissues enables users to achieve scalable manufacturing. Label-free biosensors are promising candidates for detecting cell secretomes since they can be noninvasive and do not require labor-intensive processes such as cell lysing. Moreover, most conventional monitoring techniques are single-use, conducted at the end of the fabrication process, and, challengingly, are not permissive to in-line and continual detection. To address these challenges, we developed a noninvasive and continual monitoring platform to evaluate the status of cells during the biofabrication process, with a particular focus on monitoring the transient processes that stem cells go through during in vitro differentiation over extended periods. We designed and evaluated a reusable electrochemical immunosensor with the capacity for detecting trace amounts of secreted osteogenic markers, such as osteopontin (OPN). The sensor has a low limit of detection (LOD), high sensitivity, and outstanding selectivity in complex biological media. We used this OPN immunosensor to continuously monitor on-chip osteogenesis of human mesenchymal stem cells (hMSCs) cultured 2D and 3D hydrogel constructs inside a microfluidic bioreactor for more than a month and were able to observe changing levels of OPN secretion during culture. The proposed platform can potentially be adopted for monitoring a variety of biological applications and further developed into a fully automated system for applications in advanced cellular biomanufacturing.


Subject(s)
Biosensing Techniques , Electrochemical Techniques , Lab-On-A-Chip Devices , Osteogenesis , Humans , Biosensing Techniques/methods , Biosensing Techniques/instrumentation , Electrochemical Techniques/methods , Electrochemical Techniques/instrumentation , Osteopontin/analysis , Osteopontin/metabolism , Mesenchymal Stem Cells/cytology , Immunoassay/methods , Immunoassay/instrumentation
11.
Acta Biomater ; 173: 231-246, 2024 01.
Article in English | MEDLINE | ID: mdl-38465268

ABSTRACT

Enterocutaneous fistula (ECF) is a severe medical condition where an abnormal connection forms between the gastrointestinal tract and skin. ECFs are, in most cases, a result of surgical complications such as missed enterotomies or anastomotic leaks. The constant leakage of enteric and fecal contents from the fistula site leads to skin breakdown and increases the risk of infection. Despite advances in surgical techniques and postoperative management, ECF accounts for significant mortality rates, estimated between 15-20%, and causes debilitating morbidity. Therefore, there is a critical need for a simple and effective method to seal and heal ECF. Injectable hydrogels with combined properties of robust mechanical properties and cell infiltration/proliferation have the potential to block and heal ECF. Herein, we report the development of an injectable nanoengineered adhesive hydrogel (INAH) composed of a synthetic nanosilicate (Laponite®) and a gelatin-dopamine conjugate for treating ECF. The hydrogel undergoes fast cross-linking using a co-injection method, resulting in a matrix with improved mechanical and adhesive properties. INAH demonstrates appreciable blood clotting abilities and is cytocompatible with fibroblasts. The adhesive properties of the hydrogel are demonstrated in ex vivo adhesion models with skin and arteries, where the volume stability in the hydrated internal environment facilitates maintaining strong adhesion. In vivo assessments reveal that the INAH is biocompatible, supporting cell infiltration and extracellular matrix deposition while not forming fibrotic tissue. These findings suggest that this INAH holds promising translational potential for sealing and healing ECF.


Subject(s)
Intestinal Fistula , Tissue Adhesives , Humans , Hydrogels/pharmacology , Adhesives , Gelatin , Intestinal Fistula/therapy
12.
Lab Chip ; 24(8): 2358-2359, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38501991

ABSTRACT

Correction for 'Integrated biosensors for monitoring microphysiological systems' by Lei Mou et al., Lab Chip, 2022, 22, 3801-3816, https://doi.org/10.1039/D2LC00262K.

13.
Adv Healthc Mater ; : e2302331, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38359321

ABSTRACT

Patient-derived organoids (PDOs) developed ex vivo and in vitro are increasingly used for therapeutic screening. They provide a more physiologically relevant model for drug discovery and development compared to traditional cell lines. However, several challenges remain to be addressed to fully realize the potential of PDOs in therapeutic screening. This paper summarizes recent advancements in PDO development and the enhancement of PDO culture models. This is achieved by leveraging materials engineering and microfabrication technologies, including organs-on-a-chip and droplet microfluidics. Additionally, this work discusses the application of PDOs in therapy screening to meet diverse requirements and overcome bottlenecks in cancer treatment. Furthermore, this work introduces tools for data processing and analysis of organoids, along with their microenvironment. These tools aim to achieve enhanced readouts. Finally, this work explores the challenges and future perspectives of using PDOs in drug development and personalized screening for cancer patients.

14.
J Mater Chem B ; 12(11): 2818-2830, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38411556

ABSTRACT

Personalized bone-regenerative materials have attracted substantial interest in recent years. Modern clinical settings demand the use of engineered materials incorporating patient-derived cells, cytokines, antibodies, and biomarkers to enhance the process of regeneration. In this work, we formulated short microfiber-reinforced hydrogels with platelet-rich fibrin (PRF) to engineer implantable multi-material core-shell bone grafts. By employing 3D bioprinting technology, we fabricated a core-shell bone graft from a hybrid composite hydroxyapatite-coated poly(lactic acid) (PLA) fiber-reinforced methacryolyl gelatin (GelMA)/alginate hydrogel. The overall concept involves 3D bioprinting of long bone mimic microstructures that resemble a core-shell cancellous-cortical structure, with a stiffer shell and a softer core with our engineered biomaterial. We observed a significantly enhanced stiffness in the hydrogel scaffold incorporated with hydroxyapatite (HA)-coated PLA microfibers compared to the pristine hydrogel construct. Furthermore, HA non-coated PLA microfibers were mixed with PRF and GelMA/alginate hydrogel to introduce a slow release of growth factors which can further enhance cell maturation and differentiation. These patient-specific bone grafts deliver cytokines and growth factors with distinct spatiotemporal release profiles to enhance tissue regeneration. The biocompatible and bio-responsive bone mimetic core-shell multi-material structures enhance osteogenesis and can be customized to have materials at a specific location, geometry, and material combination.


Subject(s)
Hydrogels , Osteogenesis , Humans , Hydrogels/chemistry , Durapatite , Gelatin/chemistry , Alginates/chemistry , Cytokines , Polyesters
15.
Tissue Eng Part B Rev ; 30(1): 1-14, 2024 02.
Article in English | MEDLINE | ID: mdl-37294202

ABSTRACT

Myocardial infarction results in the significant loss of cardiomyocytes (CMs) due to the ischemic injury following coronary occlusion leading to impaired contractility, fibrosis, and ultimately heart failure. Stem cell therapy emerged as a promising regenerative strategy to replenish the otherwise terminally differentiated CM to restore cardiac function. Multiple strategies have been applied to successfully differentiate diverse stem cell populations into CM-like phenotypes characterized by the expression status of signature biomarkers and observable spontaneous contractions. This article discusses the current understanding and applications of various stem cell phenotypes to drive the differentiation machinery toward CM-like lineage. Impact Statement Ischemic heart disease (IHD) extensively affects a large proportion of the population worldwide. Unfortunately, current treatments for IHD are insufficient to restore cardiac effectiveness and functionality. A growing field in regenerative cardiology explores the potential for stem cell therapy following cardiovascular ischemic episodes. The thorough understanding regarding the potential and shortcomings of translational approaches to drive versatile stem cells to cardiomyocyte lineage paves the way for multiple opportunities for next-generation cardiac management.


Subject(s)
Myocardial Infarction , Myocytes, Cardiac , Humans , Myocytes, Cardiac/metabolism , Regeneration , Myocardial Infarction/metabolism , Myocardial Infarction/therapy , Stem Cell Transplantation , Cell Differentiation
16.
Trends Biotechnol ; 42(5): 631-647, 2024 May.
Article in English | MEDLINE | ID: mdl-38158307

ABSTRACT

Electrospinning technology has garnered wide attention over the past few decades in various biomedical applications including drug delivery, cell therapy, and tissue engineering. This technology can create nanofibers with tunable fiber diameters and functionalities. However, the 2D membrane nature of the nanofibers, as well as the rigidity and low porosity of electrospun fibers, lower their efficacy in tissue repair and regeneration. Recently, new avenues have been explored to resolve the challenges associated with 2D electrospun nanofiber membranes. This review discusses recent trends in creating different electrospun nanofiber microstructures from 2D nanofiber membranes by using various post-processing methods, as well as their biotechnological applications.


Subject(s)
Biotechnology , Nanofibers , Tissue Engineering , Nanofibers/chemistry , Biotechnology/methods , Tissue Engineering/methods , Drug Delivery Systems , Humans , Biocompatible Materials/chemistry , Tissue Scaffolds/chemistry
17.
JPhys Mater ; 7(1): 012502, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38144214

ABSTRACT

This Roadmap on drug delivery aims to cover some of the most recent advances in the field of materials for drug delivery systems (DDSs) and emphasizes the role that multifunctional materials play in advancing the performance of modern DDSs in the context of the most current challenges presented. The Roadmap is comprised of multiple sections, each of which introduces the status of the field, the current and future challenges faced, and a perspective of the required advances necessary for biomaterial science to tackle these challenges. It is our hope that this collective vision will contribute to the initiation of conversation and collaboration across all areas of multifunctional materials for DDSs. We stress that this article is not meant to be a fully comprehensive review but rather an up-to-date snapshot of different areas of research, with a minimal number of references that focus upon the very latest research developments.

18.
19.
ACS Mater Au ; 3(6): 636-645, 2023 Nov 08.
Article in English | MEDLINE | ID: mdl-38089667

ABSTRACT

In recent years, fiber-based systems have been explored in the frame of tissue engineering due to their robustness in recapitulating the architecture and mechanical properties of native tissues. Such scaffolds offer anisotropic architecture capable of reproducing the native collagen fibers' orientation and distribution. Moreover, fibrous constructs might provide a biomimetic environment for cell encapsulation and proliferation as well as influence their orientation and distribution. In this work, we combine two fiber fabrication techniques, such as electrospinning and wet-spinning, in order to obtain novel cell-laden 3D fibrous layered scaffolds which can simultaneously provide: (i) mechanical support; (ii) suitable microenvironment for 3D cell encapsulation; and (iii) loading and sustained release of growth factors for promoting the differentiation of human bone marrow-derived mesenchymal stem cells (hB-MSCs). The constructs are formed from wet-spun hydrogel fibers loaded with hB-MSCs deposited on a fibrous composite electrospun matrix made of polycaprolactone, polyamide 6, and mesoporous silica nanoparticles enriched with bone morphogenetic protein-12 (BMP-12). Morphological and mechanical characterizations of the structures were carried out, and the growth factor release was assessed. The biological response in terms of cell viability, alignment, differentiation, and extracellular matrix production was investigated. Ex vivo testing of the layered structure was performed to prove the layers' integrity when subjected to mechanical stretching in the physiological range. The results reveal that 3D layered scaffolds can be proposed as valid candidates for tendon tissue engineering.

20.
Adv Drug Deliv Rev ; 203: 115142, 2023 12.
Article in English | MEDLINE | ID: mdl-37967768

ABSTRACT

As miniaturized and simplified stem cell-derived 3D organ-like structures, organoids are rapidly emerging as powerful tools for biomedical applications. With their potential for personalized therapeutic interventions and high-throughput drug screening, organoids have gained significant attention recently. In this review, we discuss the latest developments in engineering organoids and using materials engineering, biochemical modifications, and advanced manufacturing technologies to improve organoid culture and replicate vital anatomical structures and functions of human tissues. We then explore the diverse biomedical applications of organoids, including drug development and disease modeling, and highlight the tools and analytical techniques used to investigate organoids and their microenvironments. We also examine the latest clinical trials and patents related to organoids that show promise for future clinical translation. Finally, we discuss the challenges and future perspectives of using organoids to advance biomedical research and potentially transform personalized medicine.


Subject(s)
Biomedical Research , Organoids , Humans , Stem Cells , Precision Medicine/methods , Biomedical Research/methods , Drug Development
SELECTION OF CITATIONS
SEARCH DETAIL