Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.215
Filter
1.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38731971

ABSTRACT

Tendinopathy, characterized by inflammatory and degenerative changes, presents challenges in sports and medicine. In addressing the limitations of conservative management, this study focuses on developing tendon grafts using extrusion bioprinting with platelet-rich plasma (PRP)-infused hydrogels loaded with tendon cells. The objective is to understand paracrine interactions initiated by bioprinted tendon grafts in either inflamed or non-inflamed host tissues. PRP was utilized to functionalize methacrylate gelatin (GelMA), incorporating tendon cells for graft bioprinting. Bioinformatic analyses of overexpressed proteins, predictive of functional enrichment, revealed insights into PRP graft behavior in both non-inflamed and inflamed environments. PRP grafts activated inflammatory pathways, including Interleukin 17 (IL-17), neuroinflammation, Interleukin 33 (IL-33), and chemokine signaling. Interleukin 1 beta (IL-1b) in the graft environment triggered p38 mitogen-activated protein kinase (MAPK) signaling, nuclear factor kappa light chain enhancer of activated B cells (NF-kB) canonical pathway, and Vascular Endothelial Growth Factor (VEGF) signaling. Biological enrichment attributed to PRP grafts included cell chemotaxis, collagen turnover, cell migration, and angiogenesis. Acellular PRP grafts differed from nude grafts in promoting vessel length, vessel area, and junction density. Angiogenesis in cellular grafts was enhanced with newly synthesized Interleukin 8 (IL-8) in cooperation with IL-1b. In conclusion, paracrine signaling from PRP grafts, mediated by chemokine activities, influences cell migration, inflammation, and angiogenic status in host tissues. Under inflammatory conditions, newly synthesized IL-8 regulates vascularization in collaboration with PRP.


Subject(s)
Bioprinting , Platelet-Rich Plasma , Tendons , Tendons/metabolism , Bioprinting/methods , Animals , Platelet-Rich Plasma/metabolism , Humans , Tissue Engineering/methods , Hydrogels/chemistry , Tissue Scaffolds/chemistry , Tendinopathy/metabolism , Tendinopathy/therapy , Tendinopathy/pathology
2.
Arch Dermatol Res ; 316(5): 147, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38698273

ABSTRACT

Mohs Micrographic Surgery (MMS) is effective for treating common cutaneous malignancies, but complex repairs may often present challenges for reconstruction. This paper explores the potential of three-dimensional (3D) bioprinting in MMS, offering superior outcomes compared to traditional methods. 3D printing technologies show promise in advancing skin regeneration and refining surgical techniques in dermatologic surgery. A PubMed search was conducted using the following keywords: "Three-dimensional bioprinting" OR "3-D printing" AND "Mohs" OR "Mohs surgery" OR "Surgery." Peer-reviewed English articles discussing medical applications of 3D bioprinting were included, while non-peer-reviewed and non-English articles were excluded. Patients using 3D MMS models had lower anxiety scores (3.00 to 1.7, p < 0.0001) and higher knowledge assessment scores (5.59 or 93.25% correct responses), indicating better understanding of their procedure. Surgical residents using 3D models demonstrated improved proficiency in flap reconstructions (p = 0.002) and knowledge assessment (p = 0.001). Additionally, 3D printing offers personalized patient care through tailored surgical guides and anatomical models, reducing intraoperative time while enhancing surgical. Concurrently, efforts in tissue engineering and regenerative medicine are being explored as potential alternatives to address organ donor shortages, eliminating autografting needs. However, challenges like limited training and technological constraints persist. Integrating optical coherence tomography with 3D bioprinting may expedite grafting, but challenges remain in pre-printing grafts for complex cases. Regulatory and ethical considerations are paramount for patient safety, and further research is needed to understand long-term effects and cost-effectiveness. While promising, significant advancements are necessary for full utilization in MMS.


Subject(s)
Bioprinting , Mohs Surgery , Printing, Three-Dimensional , Skin Neoplasms , Humans , Bioprinting/methods , Mohs Surgery/methods , Skin Neoplasms/surgery , Tissue Engineering/methods , Models, Anatomic , Plastic Surgery Procedures/methods , Plastic Surgery Procedures/instrumentation , Surgical Flaps , Skin , Regenerative Medicine/methods
3.
Sci Rep ; 14(1): 11003, 2024 05 14.
Article in English | MEDLINE | ID: mdl-38744985

ABSTRACT

The future of organ and tissue biofabrication strongly relies on 3D bioprinting technologies. However, maintaining sterility remains a critical issue regardless of the technology used. This challenge becomes even more pronounced when the volume of bioprinted objects approaches organ dimensions. Here, we introduce a novel device called the Flexible Unique Generator Unit (FUGU), which is a unique combination of flexible silicone membranes and solid components made of stainless steel. Alternatively, the solid components can also be made of 3D printed medical-grade polycarbonate. The FUGU is designed to support micro-extrusion needle insertion and removal, internal volume adjustment, and fluid management. The FUGU was assessed in various environments, ranging from custom-built basic cartesian to sophisticated 6-axis robotic arm bioprinters, demonstrating its compatibility, flexibility, and universality across different bioprinting platforms. Sterility assays conducted under various infection scenarios highlight the FUGU's ability to physically protect the internal volume against contaminations, thereby ensuring the integrity of the bioprinted constructs. The FUGU also enabled bioprinting and cultivation of a 14.5 cm3 human colorectal cancer tissue model within a completely confined and sterile environment, while allowing for the exchange of gases with the external environment. This FUGU system represents a significant advancement in 3D bioprinting and biofabrication, paving the path toward the sterile production of implantable tissues and organs.


Subject(s)
Bioprinting , Bioreactors , Printing, Three-Dimensional , Bioprinting/methods , Humans , Tissue Engineering/methods , Sterilization , Tissue Scaffolds
4.
Biofabrication ; 16(3)2024 May 15.
Article in English | MEDLINE | ID: mdl-38697093

ABSTRACT

Organoids have emerged as crucial platforms in tissue engineering and regenerative medicine but confront challenges in faithfully mimicking native tissue structures and functions. Bioprinting technologies offer a significant advancement, especially when combined with organoid bioinks-engineered formulations designed to encapsulate both the architectural and functional elements of specific tissues. This review provides a rigorous, focused examination of the evolution and impact of organoid bioprinting. It emphasizes the role of organoid bioinks that integrate key cellular components and microenvironmental cues to more accurately replicate native tissue complexity. Furthermore, this review anticipates a transformative landscape invigorated by the integration of artificial intelligence with bioprinting techniques. Such fusion promises to refine organoid bioink formulations and optimize bioprinting parameters, thus catalyzing unprecedented advancements in regenerative medicine. In summary, this review accentuates the pivotal role and transformative potential of organoid bioinks and bioprinting in advancing regenerative therapies, deepening our understanding of organ development, and clarifying disease mechanisms.


Subject(s)
Bioprinting , Organoids , Regenerative Medicine , Tissue Engineering , Organoids/cytology , Humans , Bioprinting/methods , Tissue Engineering/methods , Animals , Regenerative Medicine/methods , Ink
5.
Jt Dis Relat Surg ; 35(2): 361-367, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38727116

ABSTRACT

OBJECTIVES: This study aims to compare the radiological, biomechanical, and histopathological results of microfracture treatment and osteochondral damage repair treatment with a new scaffold product produced by the three-dimensional (3D) bioprinting method containing gelatin-hyaluronic acid-alginate in rabbits with osteochondral damage. MATERIALS AND METHODS: A new 3D bioprinted scaffold consisting of gelatin, hyaluronic acid, and alginate designed by us was implanted into the osteochondral defect created in the femoral trochlea of 10 rabbits. By randomization, it was determined which side of 10 rabbits would be repaired with a 3D bioprinted scaffold, and microfracture treatment was applied to the other knees of the rabbits. After six months of follow-up, the rabbits were sacrificed. The results of both treatment groups were compared radiologically, biomechanically, and histopathologically. RESULTS: None of the rabbits experienced any complications. The magnetic resonance imaging evaluation showed that all osteochondral defect areas were integrated with healthy cartilage in both groups. There was no significant difference between the groups in the biomechanical load test (p=0.579). No statistically significant difference was detected in the histological examination using the modified Wakitani scores (p=0.731). CONCLUSION: Our study results showed that 3D bioprinted scaffolds exhibited comparable radiological, biomechanical, and histological properties to the conventional microfracture technique for osteochondral defect treatment.


Subject(s)
Alginates , Bioprinting , Cartilage, Articular , Gelatin , Hyaluronic Acid , Knee Joint , Printing, Three-Dimensional , Tissue Scaffolds , Animals , Rabbits , Alginates/chemistry , Gelatin/chemistry , Hyaluronic Acid/chemistry , Hyaluronic Acid/therapeutic use , Tissue Scaffolds/chemistry , Cartilage, Articular/pathology , Cartilage, Articular/injuries , Cartilage, Articular/surgery , Knee Joint/surgery , Knee Joint/pathology , Bioprinting/methods , Disease Models, Animal , Biomechanical Phenomena , Magnetic Resonance Imaging , Arthroplasty, Subchondral/methods
6.
Int J Biol Macromol ; 267(Pt 2): 131412, 2024 May.
Article in English | MEDLINE | ID: mdl-38593894

ABSTRACT

The synthesis of ideal bioceramics to guide the fate of cells and subsequent bone regeneration within the chemical, biological, and physical microenvironment is a challenging long-term task. This study developed amorphous calcium magnesium phosphate (ACMP) bioceramics via a simple co-precipitation method. The role of Mg2+ in the formation of ACMP is investigated using physicochemical and biological characterization at different Ca/Mg molar ratio of the initial reaction solution. Additionally, ACMP bioceramics show superior cytocompatibility and improved osteogenic differentiation of co-cultured MC3T3-E1 cells. Regulation of the microenvironment with Mg2+ can promote early-stage bone regeneration. For this, bioprinting technology is employed to prepare ACMP-modified 3D porous structures. Our hypothesis is that the incorporation of ACMP into methacrylated gelatin (GelMA) bioink can trigger the osteogenic differentiation of encapsulated preosteoblast and stimulate bone regeneration. The cell-laden ACMP composite structures display stable printability and superior cell viability and cell proliferation. Also, constructs loading the appropriate amount of ACMP bioceramic showed significant osteogenic differentiation activity compared to the pure GelMA. We demonstrate that the dissolved Mg2+ cation microenvironment in ACMP-modified composite constructs plays an effective biochemical role, and can regulate cell fate. Our results predict that GelMA/ACMP bioink has significant potential in patient-specific bone tissue regeneration.


Subject(s)
Bioprinting , Bone Regeneration , Calcium Phosphates , Cell Differentiation , Osteogenesis , Printing, Three-Dimensional , Tissue Scaffolds , Bone Regeneration/drug effects , Mice , Animals , Osteogenesis/drug effects , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Cell Differentiation/drug effects , Bioprinting/methods , Tissue Scaffolds/chemistry , Cell Proliferation/drug effects , Magnesium Compounds/chemistry , Magnesium Compounds/pharmacology , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Survival/drug effects , Cell Line , Tissue Engineering/methods , Osteoblasts/drug effects , Osteoblasts/cytology , Phosphates/chemistry , Phosphates/pharmacology
7.
ACS Biomater Sci Eng ; 10(5): 2616-2635, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38664996

ABSTRACT

Primary brain tumor is one of the most fatal diseases. The most malignant type among them, glioblastoma (GBM), has low survival rates. Standard treatments reduce the life quality of patients due to serious side effects. Tumor aggressiveness and the unique structure of the brain render the removal of tumors and the development of new therapies challenging. To elucidate the characteristics of brain tumors and examine their response to drugs, realistic systems that mimic the tumor environment and cellular crosstalk are desperately needed. In the past decade, 3D GBM models have been presented as excellent platforms as they allowed the investigation of the phenotypes of GBM and testing innovative therapeutic strategies. In that scope, 3D bioprinting technology offers utilities such as fabricating realistic 3D bioprinted structures in a layer-by-layer manner and precisely controlled deposition of materials and cells, and they can be integrated with other technologies like the microfluidics approach. This Review covers studies that investigated 3D bioprinted brain tumor models, especially GBM using 3D bioprinting techniques and essential parameters that affect the result and quality of the study like frequently used cells, the type and physical characteristics of hydrogel, bioprinting conditions, cross-linking methods, and characterization techniques.


Subject(s)
Bioprinting , Brain Neoplasms , Glioblastoma , Printing, Three-Dimensional , Humans , Glioblastoma/pathology , Bioprinting/methods , Brain Neoplasms/pathology , Animals , Brain/pathology , Tissue Engineering/methods
8.
Biomed Mater ; 19(3)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38626774

ABSTRACT

Bioinks play a crucial role in tissue engineering, influencing mechanical and chemical properties of the printed scaffold as well as the behavior of encapsulated cells. Recently, there has been a shift from animal origin materials to their synthetic alternatives. In this context, we present here bioinks based on fully synthetic and biodegradable poly(α,L-amino acids) (PolyAA) as an alternative to animal-based gelatin methacrylate (Gel-Ma) bioinks. Additionally, we first reported the possibility of the visible light photoinitiated incorporation of the bifunctional cell adhesive RGD peptide into the PolyAA hydrogel matrix. The obtained hydrogels are shown to be cytocompatible, and their mechanical properties closely resemble those of gelatin methacrylate-based scaffolds. Moreover, combining the unique properties of PolyAA-based bioinks, the photocrosslinking strategy, and the use of droplet-based printing allows the printing of constructs with high shape fidelity and structural integrity from low-viscosity bioinks without using any sacrificial components. Overall, presented PolyAA-based materials are a promising and versatile toolbox that extends the range of bioinks for droplet bioprinting.


Subject(s)
Amino Acids , Biocompatible Materials , Gelatin , Hydrogels , Light , Tissue Engineering , Tissue Scaffolds , Hydrogels/chemistry , Tissue Scaffolds/chemistry , Tissue Engineering/methods , Gelatin/chemistry , Amino Acids/chemistry , Biocompatible Materials/chemistry , Animals , Bioprinting/methods , Oligopeptides/chemistry , Ink , Methacrylates/chemistry , Humans , Printing, Three-Dimensional , Materials Testing , Mice , Viscosity
9.
Biofabrication ; 16(3)2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38569491

ABSTRACT

Regenerative healing of spinal cord injury (SCI) poses an ongoing medical challenge by causing persistent neurological impairment and a significant socioeconomic burden. The complexity of spinal cord tissue presents hurdles to successful regeneration following injury, due to the difficulty of forming a biomimetic structure that faithfully replicates native tissue using conventional tissue engineering scaffolds. 3D bioprinting is a rapidly evolving technology with unmatched potential to create 3D biological tissues with complicated and hierarchical structure and composition. With the addition of biological additives such as cells and biomolecules, 3D bioprinting can fabricate preclinical implants, tissue or organ-like constructs, andin vitromodels through precise control over the deposition of biomaterials and other building blocks. This review highlights the characteristics and advantages of 3D bioprinting for scaffold fabrication to enable SCI repair, including bottom-up manufacturing, mechanical customization, and spatial heterogeneity. This review also critically discusses the impact of various fabrication parameters on the efficacy of spinal cord repair using 3D bioprinted scaffolds, including the choice of printing method, scaffold shape, biomaterials, and biological supplements such as cells and growth factors. High-quality preclinical studies are required to accelerate the translation of 3D bioprinting into clinical practice for spinal cord repair. Meanwhile, other technological advances will continue to improve the regenerative capability of bioprinted scaffolds, such as the incorporation of nanoscale biological particles and the development of 4D printing.


Subject(s)
Bioprinting , Printing, Three-Dimensional , Spinal Cord Injuries , Tissue Scaffolds , Spinal Cord Injuries/therapy , Bioprinting/methods , Humans , Animals , Tissue Scaffolds/chemistry , Tissue Engineering , Biocompatible Materials/chemistry
10.
J Biomech Eng ; 146(9)2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38557592

ABSTRACT

Development of respiratory tissue constructs is challenging due to the complex structure of native respiratory tissue and the unique biomechanical conditions induced by breathing. While studies have shown that the inclusion of biomechanical stimulus mimicking physiological conditions greatly benefits the development of engineered tissues, to our knowledge no studies investigating the influence of biomechanical stimulus on the development of respiratory tissue models produced through three-dimensional (3D) bioprinting have been reported. This paper presents a study on the utilization of a novel breath-mimicking ventilated incubator to impart biomechanical stimulus during the culture of 3D respiratory bioprinted constructs. Constructs were bioprinted using an alginate/collagen hydrogel containing human primary pulmonary fibroblasts with further seeding of human primary bronchial epithelial cells. Biomechanical stimulus was then applied via a novel ventilated incubator capable of mimicking the pressure and airflow conditions of multiple breathing conditions: standard incubation, shallow breathing, normal breathing, and heavy breathing, over a two-week time period. At time points between 1 and 14 days, constructs were characterized in terms of mechanical properties, cell proliferation, and morphology. The results illustrated that incubation conditions mimicking normal and heavy breathing led to greater and more continuous cell proliferation and further indicated a more physiologically relevant respiratory tissue model.


Subject(s)
Bioprinting , Tissue Scaffolds , Humans , Tissue Scaffolds/chemistry , Tissue Engineering/methods , Hydrogels/chemistry , Respiration , Printing, Three-Dimensional , Bioprinting/methods
11.
Biofabrication ; 16(3)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38574552

ABSTRACT

The advent of 3D bioprinting technologies in tissue engineering has unlocked the potential to fabricatein vitrotissue models, overcoming the constraints associated with the shape limitations of preformed scaffolds. However, achieving an accurate mimicry of complex tissue microenvironments, encompassing cellular and biochemical components, and orchestrating their supramolecular assembly to form hierarchical structures while maintaining control over tissue formation, is crucial for gaining deeper insights into tissue repair and regeneration. Building upon our expertise in developing competent three-dimensional tissue equivalents (e.g. skin, gut, cervix), we established a two-step bottom-up approach involving the dynamic assembly of microtissue precursors (µTPs) to generate macroscopic functional tissue composed of cell-secreted extracellular matrix (ECM). To enhance precision and scalability, we integrated extrusion-based bioprinting technology into our established paradigm to automate, control and guide the coherent assembly ofµTPs into predefined shapes. Compared to cell-aggregated bioink, ourµTPs represent a functional unit where cells are embedded in their specific ECM.µTPs were derived from human dermal fibroblasts dynamically seeded onto gelatin-based microbeads. After 9 days,µTPs were suspended (50% v/v) in Pluronic-F127 (30% w/v) (µTP:P30), and the obtained formulation was loaded as bioink into the syringe of the Dr.INVIVO-4D6 extrusion based bioprinter.µTP:P30 bioink showed shear-thinning behavior and temperature-dependent viscosity (gel atT> 30 °C), ensuringµTPs homogenous dispersion within the gel and optimal printability. The bioprinting involved extruding several geometries (line, circle, and square) into Pluronic-F127 (40% w/v) (P40) support bath, leveraging its shear-recovery property. P40 effectively held the bioink throughout and after the bioprinting procedure, untilµTPs fused into a continuous connective tissue.µTPs fusion dynamics was studied over 8 days of culture, while the resulting endogenous construct underwent 28 days culture. Histological, immunofluorescence analysis, and second harmonic generation reconstruction revealed an increase in endogenous collagen and fibronectin production within the bioprinted construct, closely resembling the composition of the native connective tissues.


Subject(s)
Bioprinting , Polyethylenes , Polypropylenes , Tissue Scaffolds , Humans , Tissue Scaffolds/chemistry , Bioprinting/methods , Poloxamer , Uridine Triphosphate , Tissue Engineering/methods , Printing, Three-Dimensional
12.
Nat Commun ; 15(1): 3565, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38670999

ABSTRACT

Bioprinting that can synchronously deposit cells and biomaterials has lent fresh impetus to the field of tissue regeneration. However, the unavoidable occurrence of cell damage during fabrication process and intrinsically poor mechanical stability of bioprinted cell-laden scaffolds severely restrict their utilization. As such, on basis of heart-inspired hollow hydrogel-based scaffolds (HHSs), a mechanical-assisted post-bioprinting strategy is proposed to load cells into HHSs in a rapid, uniform, precise and friendly manner. HHSs show mechanical responsiveness to load cells within 4 s, a 13-fold increase in cell number, and partitioned loading of two types of cells compared with those under static conditions. As a proof of concept, HHSs with the loading cells show an enhanced regenerative capability in repair of the critical-sized segmental and osteoporotic bone defects in vivo. We expect that this post-bioprinting strategy can provide a universal, efficient, and promising way to promote cell-based regenerative therapy.


Subject(s)
Bioprinting , Bone Regeneration , Hydrogels , Tissue Engineering , Tissue Scaffolds , Animals , Tissue Scaffolds/chemistry , Hydrogels/chemistry , Bioprinting/methods , Tissue Engineering/methods , Humans , Bone and Bones , Mice , Mesenchymal Stem Cells/cytology , Biocompatible Materials/chemistry , Osteoporosis/therapy
13.
Int J Biol Macromol ; 266(Pt 1): 131281, 2024 May.
Article in English | MEDLINE | ID: mdl-38641503

ABSTRACT

As an emerging new manufacturing technology, Three-dimensional (3D) bioprinting provides the potential for the biomimetic construction of multifaceted and intricate architectures of functional integument, particularly functional biomimetic dermal structures inclusive of cutaneous appendages. Although the tissue-engineered skin with complete biological activity and physiological functions is still cannot be manufactured, it is believed that with the advances in matrix materials, molding process, and biotechnology, a new generation of physiologically active skin will be born in the future. In pursuit of furnishing readers and researchers involved in relevant research to have a systematic and comprehensive understanding of 3D printed tissue-engineered skin, this paper furnishes an exegesis on the prevailing research landscape, formidable obstacles, and forthcoming trajectories within the sphere of tissue-engineered skin, including: (1) the prevalent biomaterials (collagen, chitosan, agarose, alginate, etc.) routinely employed in tissue-engineered skin, and a discerning analysis and comparison of their respective merits, demerits, and inherent characteristics; (2) the underlying principles and distinguishing attributes of various current printing methodologies utilized in tissue-engineered skin fabrication; (3) the present research status and progression in the realm of tissue-engineered biomimetic skin; (4) meticulous scrutiny and summation of the extant research underpinning tissue-engineered skin inform the identification of prevailing challenges and issues.


Subject(s)
Biocompatible Materials , Bioprinting , Printing, Three-Dimensional , Skin , Tissue Engineering , Tissue Engineering/methods , Bioprinting/methods , Humans , Biocompatible Materials/chemistry , Animals , Tissue Scaffolds/chemistry , Skin, Artificial
14.
Biofabrication ; 16(3)2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38565131

ABSTRACT

Extrusion-based bioprinting is a promising technology for the fabrication of complex three-dimensional (3D) tissue-engineered constructs. To further improve the printing accuracy and provide mechanical support during the printing process, hydrogel-based support bath materials have been developed. However, the gel structure of some support bath materials can be compromised when exposed to certain bioink crosslinking cues, hence their compatibility with bioinks can be limited. In this study, a xanthan gum-based composite support material compatible with multiple crosslinking mechanisms is developed. Different support bath materials can have different underlying polymeric structures, for example, particulate suspensions and polymer solution with varying supramolecular structure) and these properties are governed by a variety of different intermolecular interactions. However, common rheological behavior can be expected because they have similar demonstrated performance and functionality. To provide a detailed exploration/identification of the common rheological properties expressed by different support bath materials from a unified perspective, benchmark support bath materials from previous studies were prepared. A comparative rheological study revealed both the structural and shear behavior characteristics shared by support bath materials, including yield stress, gel complex moduli, shear-thinning behavior, and self-healing properties. Gel structural stability and functionality of support materials were tested in the presence of various crosslinking stimuli, confirming the versatility of the xanthan-based support material. We further investigated the effect of support materials and the diameter of extrusion needles on the printability of bioinks to demonstrate the improvement in bioink printability and structural integrity. Cytotoxicity and cell encapsulation viability tests were carried out to confirm the cell compatibility of the xanthan gum-based support bath material. We propose and demonstrate the versatility and compatibility of the novel support bath material and provide detailed new insight into the essential properties and behavior of these materials that serve as a guide for further development of support bath-based 3D bioprinting.


Subject(s)
Bioprinting , Tissue Engineering , Polysaccharides, Bacterial , Rheology , Printing, Three-Dimensional , Bioprinting/methods , Hydrogels/chemistry , Tissue Scaffolds/chemistry
15.
Stem Cell Res Ther ; 15(1): 101, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38589956

ABSTRACT

In response to the growing demand for organ substitutes, tissue engineering has evolved significantly. However, it is still challenging to create functional tissues and organs. Tissue engineering from the 'bottom-up' is promising on solving this problem due to its ability to construct tissues with physiological complexity. The workflow of this strategy involves two key steps: the creation of building blocks, and the subsequent assembly. There are many techniques developed for the two pivotal steps. Notably, bioprinting is versatile among these techniques and has been widely used in research. With its high level of automation, bioprinting has great capacity in engineering tissues with precision and holds promise to construct multi-material tissues. In this review, we summarize the techniques applied in fabrication and assembly of building blocks. We elaborate mechanisms and applications of bioprinting, particularly in the 'bottom-up' strategy. We state our perspectives on future trends of bottom-up tissue engineering, hoping to provide useful reference for researchers in this field.


Subject(s)
Bioprinting , Bioprinting/methods , Tissue Engineering/methods , Tissue Scaffolds
16.
Biomater Adv ; 160: 213850, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38626580

ABSTRACT

Decellularized extracellular matrix (dECM) is an excellent natural source for 3D bioprinting materials due to its inherent cell compatibility. In vat photopolymerization, the use of dECM-based bioresins is just emerging, and extensive research is needed to fully exploit their potential. In this study, two distinct methacryloyl-functionalized, photocrosslinkable dECM-based bioresins were prepared from digested porcine liver dECM through functionalization with glycidyl methacrylate (GMA) or conventional methacrylic anhydride (MA) under mild conditions for systematic comparison. Although the chemical modifications did not significantly affect the structural integrity of the dECM proteins, mammalian cells encapsulated in the respective hydrogels performed differently in long-term culture. In either case, photocrosslinking during 3D (bio)printing resulted in transparent, highly swollen, and soft hydrogels with good shape fidelity, excellent biomimetic properties and tunable mechanical properties (~ 0.2-2.5 kPa). Interestingly, at a similar degree of functionalization (DOF ~ 81.5-83.5 %), the dECM-GMA resin showed faster photocrosslinking kinetics in photorheology resulting in lower final stiffness and faster enzymatic biodegradation compared to the dECM-MA gels, yet comparable network homogeneity as assessed via Brillouin imaging. While human hepatic HepaRG cells exhibited comparable cell viability directly after 3D bioprinting within both materials, cell proliferation and spreading were clearly enhanced in the softer dECM-GMA hydrogels at a comparable degree of crosslinking. These differences were attributed to the additional hydrophilicity introduced to dECM via methacryloylation through GMA compared to MA. Due to its excellent printability and cytocompatibility, the functional porcine liver dECM-GMA biomaterial enables the advanced biofabrication of soft 3D tissue analogs using vat photopolymerization-based bioprinting.


Subject(s)
Extracellular Matrix , Hydrogels , Methacrylates , Polymerization , Animals , Methacrylates/chemistry , Swine , Hydrogels/chemistry , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Liver , Humans , Printing, Three-Dimensional , Photochemical Processes , Bioprinting/methods , Biocompatible Materials/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Cross-Linking Reagents/chemistry , Epoxy Compounds/chemistry
17.
Biosci Rep ; 44(5)2024 May 29.
Article in English | MEDLINE | ID: mdl-38563479

ABSTRACT

Osteoarthritis (OA) is a long-term, persistent joint disorder characterized by bone and cartilage degradation, resulting in tightness, pain, and restricted movement. Current attempts in cartilage regeneration are cell-based therapies using stem cells. Multipotent stem cells, such as mesenchymal stem cells (MSCs), and pluripotent stem cells, such as embryonic stem cells (ESCs), have been used to regenerate cartilage. However, since the discovery of human-induced pluripotent stem cells (hiPSCs) in 2007, it was seen as a potential source for regenerative chondrogenic therapy as it overcomes the ethical issues surrounding the use of ESCs and the immunological and differentiation limitations of MSCs. This literature review focuses on chondrogenic differentiation and 3D bioprinting technologies using hiPSCS, suggesting them as a viable source for successful tissue engineering. METHODS: A literature search was conducted using scientific search engines, PubMed, MEDLINE, and Google Scholar databases with the terms 'Cartilage tissue engineering' and 'stem cells' to retrieve published literature on chondrogenic differentiation and tissue engineering using MSCs, ESCs, and hiPSCs. RESULTS: hiPSCs may provide an effective and autologous treatment for focal chondral lesions, though further research is needed to explore the potential of such technologies. CONCLUSIONS: This review has provided a comprehensive overview of these technologies and the potential applications for hiPSCs in regenerative medicine.


Subject(s)
Cell Differentiation , Chondrogenesis , Induced Pluripotent Stem Cells , Tissue Engineering , Humans , Tissue Engineering/methods , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Animals , Osteoarthritis/therapy , Osteoarthritis/pathology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Regenerative Medicine/methods , Cartilage/metabolism , Cartilage/cytology , Bioprinting/methods , Printing, Three-Dimensional , Regeneration
18.
Stem Cells Transl Med ; 13(5): 425-435, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38502194

ABSTRACT

The ultimate goal of cardiac tissue engineering is to generate new muscle to repair or replace the damaged heart. This requires advances in stem cell technologies to differentiate billions of cardiomyocytes, together with advanced biofabrication approaches such as 3D bioprinting to achieve the requisite structure and contractile function. In this concise review, we cover recent progress in 3D bioprinting of cardiac tissue using pluripotent stem cell-derived cardiomyocytes, key design criteria for engineering aligned cardiac tissues, and ongoing challenges in the field that must be addressed to realize this goal.


Subject(s)
Bioprinting , Myocytes, Cardiac , Printing, Three-Dimensional , Tissue Engineering , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Humans , Tissue Engineering/methods , Bioprinting/methods , Animals , Cell Differentiation , Pluripotent Stem Cells/cytology , Tissue Scaffolds/chemistry , Myocardium/cytology , Myocardium/metabolism
19.
Adv Sci (Weinh) ; 11(18): e2308276, 2024 May.
Article in English | MEDLINE | ID: mdl-38514919

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) continuously generate platelets throughout one's life. Inherited Platelet Disorders affect ≈ 3 million individuals worldwide and are characterized by defects in platelet formation or function. A critical challenge in the identification of these diseases lies in the absence of models that facilitate the study of hematopoiesis ex vivo. Here, a silk fibroin-based bioink is developed and designed for 3D bioprinting. This bioink replicates a soft and biomimetic environment, enabling the controlled differentiation of HSPCs into platelets. The formulation consisting of silk fibroin, gelatin, and alginate is fine-tuned to obtain a viscoelastic, shear-thinning, thixotropic bioink with the remarkable ability to rapidly recover after bioprinting and provide structural integrity and mechanical stability over long-term culture. Optical transparency allowed for high-resolution imaging of platelet generation, while the incorporation of enzymatic sensors allowed quantitative analysis of glycolytic metabolism during differentiation that is represented through measurable color changes. Bioprinting patient samples revealed a decrease in metabolic activity and platelet production in Inherited Platelet Disorders. These discoveries are instrumental in establishing reference ranges for classification and automating the assessment of treatment responses. This model has far-reaching implications for application in the research of blood-related diseases, prioritizing drug development strategies, and tailoring personalized therapies.


Subject(s)
Bioprinting , Blood Platelets , Cell Differentiation , Fibroins , Hematopoiesis , Printing, Three-Dimensional , Fibroins/metabolism , Fibroins/chemistry , Bioprinting/methods , Humans , Blood Platelets/metabolism , Hematopoiesis/physiology , Ink , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Gelatin/chemistry
20.
Biofabrication ; 16(2)2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38447217

ABSTRACT

As bioprinting advances into clinical relevance with patient-specific tissue and organ constructs, it must be capable of multi-material fabrication at high resolutions to accurately mimick the complex tissue structures found in the body. One of the most fundamental structures to regenerative medicine is microvasculature. Its continuous hierarchical branching vessel networks bridge surgically manipulatable arteries (∼1-6 mm) to capillary beds (∼10µm). Microvascular perfusion must be established quickly for autologous, allogeneic, or tissue engineered grafts to survive implantation and heal in place. However, traditional syringe-based bioprinting techniques have struggled to produce perfusable constructs with hierarchical branching at the resolution of the arterioles (∼100-10µm) found in microvascular tissues. This study introduces the novel CEVIC bioprinting device (i.e.ContinuouslyExtrudedVariableInternalChanneling), a multi-material technology that breaks the current extrusion-based bioprinting paradigm of pushing cell-laden hydrogels through a nozzle as filaments, instead, in the version explored here, extruding thin, wide cell-laden hydrogel sheets. The CEVIC device adapts the chaotic printing approach to control the width and number of microchannels within the construct as it is extruded (i.e. on-the-fly). Utilizing novel flow valve designs, this strategy can produce continuous gradients varying geometry and materials across the construct and hierarchical branching channels with average widths ranging from 621.5 ± 42.92%µm to 11.67 ± 14.99%µm, respectively, encompassing the resolution range of microvascular vessels. These constructs can also include fugitive/sacrificial ink that vacates to leave demonstrably perfusable channels. In a proof-of-concept experiment, a co-culture of two microvascular cell types, endothelial cells and pericytes, sustained over 90% viability throughout 1 week in microchannels within CEVIC-produced gelatin methacryloyl-sodium alginate hydrogel constructs. These results justify further exploration of generating CEVIC-bioprinted microvasculature, such as pre-culturing and implantation studies.


Subject(s)
Bioprinting , Endothelial Cells , Humans , Bioprinting/methods , Tissue Engineering/methods , Hydrogels/chemistry , Printing, Three-Dimensional , Tissue Scaffolds/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...