IOP Publishing

Biofabrication

Published by IOP Publishing and International Society for Biofabrication (ISBF).

Online ISSN: 1758-5090

·

Print ISSN: 1758-5082

Journal websiteAuthor guidelines

Top-read articles

129 reads in the past 30 days

The basic 3D printing process includes six steps: concept generation, 3D CAD design, STL file creation, G-code conversion, 3D printing, and processing/evaluation.
Schematic illustration of fused deposition modeling (FDM) 3D printer consisting of nozzle, heated liquefier, roller, heating platform, and rotary filament table.
Schematic illustration of four vat photopolymerization 3D printing processes: (A) bottom-up stereolithography (SLA), (B) top-down SLA, (C) digital light processing (DLP), and (D) continuous liquid interface production (CLIP).
Schematic illustration of selective laser sintering (SLS), which is a subtechnique of powder bed fusion 3D printing.
Schematic illustration of jet printing (JTP).

+9

3D printing processes in precise drug delivery for personalized medicine

April 2024

·

614 Reads

·

30 Citations

·

Bo Han

·

·

[...]

·

Download

Aims and scope


Biofabrication focuses on cutting-edge research regarding the use of cells, proteins, biological materials and biomaterials as building blocks to manufacture biological systems and/or therapeutic products. Emphasis is on the development of fabrication technologies, modelling of the fabricated constructs and maturation of biofabricated objects towards the intended tissue types. It includes the following topics- Cell, tissue and organ printing, patterning and assembly, Biofabricated cell/biological material-integrated systems and medical devices, Novel 3D tissue scaffold fabrication, Integrated bio- and micro/nano-fabrication, Protein/biomolecule printing, patterning and assembly, Modelling of the biofabrication processes and biofabricated constructs

Recent articles


Genetically modified cell membrane proteins in tissue engineering and regenerative medicine
  • Literature Review
  • Publisher preview available

May 2025

·

27 Reads

Genetically modified cell membrane proteins can effectively regulate cell proliferation and differentiation, while also integrating novel biomaterials. As a promising biomedical tool, this technology has broad applications in tissue engineering and regenerative medicine. Both viral and non-viral gene transfection methods have been employed to create genetically modified cell membrane proteins. Numerous studies have demonstrated the significant efficacy of genetically modified cell membrane proteins in promoting bone regeneration, treating cardiovascular diseases, aiding lung injury recovery, advancing immunotherapy, and in applications involving engineered cell membrane sheets and cell spheroids. However, this technology faces several limitations, including biosafety and ethical concerns associated with genetic modification. This article summarizes recent advances in genetically modified cell membrane proteins, detailing their preparation, applications, limitations, and future directions.


Multifunctional nanoplatform based on polyethylene glycol-folic acid modified UiO-66 (Zr) as drug delivery platform for enhanced therapy of cancer

May 2025

Oral squamous cell carcinoma (OSCC) is the most common malignant tumor in the head and neck. Due to low bioavailability and passive targetability of anticancer drugs show great limitations in cancer therapy, the treatment of OSCC faces major challenges. Folic acid (FA) targeting can deliver anticancer drugs efficiently into the tumor environment, further enhance the anti-cancer efficacy. Herein, the nanoplatform based on UiO-66 that encapsulated with an effective FA targeting ligands and the pH-responsive polyethylene glycol (PEG) layer for the targeted delivery of berberine (Ber) is constructed for fighting against OSCC. The FA modification and controlled pH-responsiveness enable the targeted delivery of UiO-66/PEG-FA, which promotes the release of Ber and increases the cumulative intracellular Ber concentration, which both promote consumption of glutathione (GSH) and induced generation of reactive oxygen species (ROS), further stimulate the secretion of inflammatory factors (TNF-α and IL-1β). A comprehensive evaluation of in vitro and in vivo experiments show that UiO-66@Ber/PEG-FA promote autophagy and apoptosis of tumor cells by regulating the expression of Beclin-1, ATG13, BAX and Bcl-2, and effectively inhibit tumor growth. Overall, UiO-66@Ber/PEG-FA exhibit superior pH-responsiveness and targeted therapeutic efficiencies in vitro and vivo, it can serve as an approach for OSCC therapy.


Method for production and bioprinting of PAMs (a) cellular collagen solution, alginate solution, and CaCl2 solution are combined in a spinning device to form a solid core-shell fiber, which is wound on a motorized spool. (b) The spool is detached from the motor and the immature fiber is cultured in a dish to form an aligned cellular fiber. (c) The mature fiber is removed from the spool and laid in a cutting device, where it is cut into consistent fragments. (d) Alginate lyase removes the shell from the fragments, releasing the PAMs. (e) PAMs are combined with hydrogel bioink and extruded to lay down the PAMs in alignment with the printing direction. (f) A simplified depiction of the complexity of smooth muscle fascicle alignment at the GEJ, highlighting the non-orthogonal nature of the alignment of the key fascicle groups required for normal valve function.
Characteristics of the fiber spinning system. (a) Drawing of the spinning device with the inset image highlighting the two spinnerets in series. (b) A micro-CT scan of the 3D printed spinning device showing one of the internal spinnerets. (c) The micro-CT is compared to the original computer-aided design, demonstrating faithful reproduction of the modeled geometry with interior dimensions slightly narrower than designed, including at key diameters in the spinneret region. (d) The total diameter of the spun fiber increased with the fiber flow rate and decreased with the sheath flow rate. (e) The fiber cross-section depended on the ratio of core and shell flow rates. (f) Erioglaucine dye did not affect measured ATP at or below concentrations of 100 µM (n = 3). (g) When maintained at 4 °C, the core collagen solution has at least 30 min of working time for the spinning process and quickly forms a gel when warmed to room temperature. (h)–(k) Adding the erioglaucine dye to the alginate shell greatly increased the visibility of the fiber during the spinning process. Arrows point to the fiber where it can be seen exiting the surface of the crosslinking bath. (h) No dye, (i) 10 µM, (j) 100 µM, (k) 1000 µM. (l) A microscopic view of the submerged tip of the microfluidic device shows the core-shell fiber being carried out by a stream of calcium chloride. (m) Using a cell concentration of 10 million cells per ml, the 4 mg ml⁻¹ collagen gel core resulted in an unstable fiber that began breaking into clusters (arrows) by day 7, unlike fibers made with (n) 5 mg ml⁻¹ collagen cores which maintained their integrity. (o) The lengths of fiber output and corresponding theoretical numbers of 2 mm microtissues that can be produced from a volumetric input into the core channel of the microfluidic device are shown. For all panels, an asterisk (*) represents p < 0.05. Scale bars represent (l) 2 mm and (m, n) 500 µm.
Protein expression and cell alignment during smooth muscle fiber development. (a) Epifluorescence microscopy of the fibers for key markers of smooth muscle cells (SM-MHC, Cx43, and calponin-1) with nuclear counterstains. (b) The normalized expression of the genes (MYH11, CNN1, GJA1) encoding the markers in panel (a) was measured by qPCR and normalized to day 0 expression (n = 4). (c) Quantification of the cell alignment within fibers and unaligned controls is shown as a probability density plot, with data extracted from confocal microscopy of phalloidin 488 on day 7 (representative images in panels (d)–(e), n = 10 regions of interest, error bars depicted as shaded region show standard deviation). False-colored phalloidin 488 labeling shows the alignment of actin fibers with color-coded directionality, demonstrating cytoskeletal alignment in the (d) aligned fibers but not in the (e) unaligned control. Color-matched legends are shown in the lower left corners of panels (d)–(e), and representative regions of interest used for the quantification in (c) are plotted as white boxes. Scale bars = 100 µm for all panels.
Properties of the PAMs after processing. (a) The cutting guide with a razor produces fibers of different lengths depending on the nominal cutting intervals (n = 30). (b) After cutting, releasing the PAMs from the alginate shell results in contraction, which can be partially ameliorated by cytochalasin D (actin polymerization inhibitor) or Y-27 632 (ROCK inhibitor) (n = 11–12). Representative images of (c) PAMS before release and (d) Y-27 632-treated PAMs after release. (e) Qualitative imaging with a combination of brightfield, ethidium homodimer (red), and calcein AM (green) of PAM before release from the alginate shell shows cell death at the cut end. Representative images of (f) methanol-treated dead control and (g) cut fiber end (cut end at the top of image) were used for quantification. (h) Quantified LIVE-DEAD staining shows that the cutting process results in moderately increased cell death near the cut compared to the center region of the PAMs. (i) PAMs were bioprinted into circular patterns to demonstrate the alignment capability of the printing method. The phalloidin 488 labeled tissues were then processed with orientationJ to produce false-color images (j) where color indicates alignment direction. An inset image in the lower right of panel j shows the orientationJ color-coding applied to a set of perfect circles, showing how panel j would appear if the microtissues were perfectly aligned with the circular print paths (e.g. elements that are oriented horizontally are coded as cyan, vertical elements are coded red, etc.). The red and blue boxes denote the regions of interest used for alignment quantification. (k) Shows the plots of the mean alignments produced (n = 9), with alignment probability as a function of the angle. The standard deviation is plotted as a shaded region around the mean curve. A theoretical uniform distribution is plotted as an example of a purely unaligned distribution. As expected, the orthogonal ROIs produce complementary probability curves aligning generally at 0° or ±90°. (l) A photograph showing a GEJ-like construct printed onto a scaffold. The gross alignment of the microtissues can be seen in the printed structure. (m) Manual false coloring was added to the photograph to highlight the microtissues in the three printing regions. The inset image depicts the three key fiber directions found in a normal GEJ. Scale bars represent (c), (d) 500 µm, (e), (f), (g) 200 µm, (i, j) 1 mm, and (l, m) 1 mm. Asterisks represent p < 0.05 (*), p < 0.01(**), p < 0.001 (***) and p < 0.0001 (****).
Efficient wet-spinning of pre-aligned microtissues for 3D bioprinting complex tissue alignment

Engineering functional smooth muscle tissues requires precise control of cellular alignment, particularly in complex anatomical regions such as the gastroesophageal junction (GEJ). We present a scalable wet-spinning approach for generating pre-aligned microtissues (PAMs) from immortalized human esophageal smooth muscle cells embedded in a collagen-alginate core-shell fiber. After maturation, fibers were sectioned into uniform PAMs with preserved alignment and high cell viability. Immunofluorescence and gene expression analyses confirmed the expression of key contractile markers. PAMs were incorporated into a gelatin-methacryloyl bioink and 3D bioprinted to demonstrate alignment along the extrusion path. This method does not require specialized culture platforms and enables efficient production of aligned microtissues for bioprinting. It offers a promising strategy for fabricating anisotropic tissues and may facilitate the reconstruction of complex muscle structures such as the GEJ.


Additive manufacturing of silicon nitride fiber-reinforced polyetheretherketone composites with enhanced mechanical strength and multifunctional bioactivity for load-bearing bone defect repair

May 2025

Polyetheretherketone (PEEK) is increasingly applied in bone defect repair due to its excellent biocompatibility and absence of artifact formation. However, the bio-inertness and inadequate mechanical properties of untreated PEEK remain significant challenges for PEEK-based implants. Hence, this study prepares a series of silicon nitride (Si3N4) fiber-reinforced PEEK composite porous scaffolds using twin-screw melt mixing-extrusion and material extrusion 3D printing. Comprehensive evaluations assess the mechanical properties, biocompatibility, osteogenic differentiation, angiogenesis activities, and antibacterial performances of various composites. Characterization results show that Si3N4 fiber-reinforced PEEK composites exhibit excellent printability, with well-oriented Si3N4 fibers uniformly distributed throughout the matrix. Furthermore, compared to non-reinforced PEEK, the addition of 8% Si₃N₄ fibers enhanced Young's modulus by 52.2% (6.36 GPa). Additionally, both in vitro and in vivo results indicate that all composite scaffolds exhibit excellent biocompatibility. Notably, the 8% Si₃N₄ fiber-reinforced PEEK composite demonstrated optimal multifunctional performance in osteogenic induction, angiogenic capacity, and antibacterial efficacy, significantly outperforming other experimental groups. In conclusion, this study offers a solution for enhancing the mechanical, anti-infective, and osseointegrative properties of PEEK, demonstrating its great potential for expanding the application of non-metallic orthopedic implants in bone defect repair.


Advances in microfluidic biofabrication technology for bone metastasis modeling

May 2025

·

3 Reads

Studying bone metastasis in in vitro models is essential for understanding the mechanisms driving this process, developing effective therapeutic strategies, and evaluating potential treatments for metastatic cancer patients. To this end, traditional two-dimensional (2D) cell culture models fail to replicate the native three-dimensional (3D) tissue microenvironment, resulting in significant disparities in biologically relevant behaviors and drug responses. The shift from 2D to 3D cell culture techniques represents an important step toward creating more biomimetic bone metastasis models. These systems more effectively emulate and replicate the complex interactions between cancer cells and bone tissue, including essential cell-cell and cell-extracellular matrix interactions, as well as in vivo biomechanical cues. The development and application of microfluidic-based 3D cancer models, incorporating diverse shapes, architectures, and modular structures such as organ-on-chip platforms, enable comprehensive screening and exploration of cellular interplay, the dissection of signaling pathways, and the resolution of limitations associated with traditional models. This review highlights recent advancements in microfluidic-based 3D bone metastasis models and examines innovative applications of this technology. These include hydrogel-based spherical and filaments biofabrication approaches, 2D and 3D tumor on-a- chips, and drug screening techniques such as concentration gradient generator-based, microdroplet-based, and microarray-based chips, as well as tumor tissue chips. Additionally, we discuss the benefits and limitations of these approaches in treating bone metastases and propose future directions for advancing microfluidic platforms in drug discovery and this research field.


High resolution melt electro-written scaffolds promote alignment of human skeletal muscle cells

May 2025

·

2 Reads

Advanced tissue engineering strategies are vital to address challenging musculoskeletal conditions, such as volumetric muscle loss. These disorders impose a considerable economic burden and affect individuals' quality of life, highlighting the need for innovative treatments, such as tissue engineering, to address these challenges. Here, we examine how scaffold fibre orientation influences mechanical properties and cellular behaviour by utilising Melt Electrowriting (MEW) as a high-resolution 3D printing technique that combines aspects of electrospinning and melt based polymer deposition. In this work, we investigated the effects of fibre orientation in MEW scaffolds, and its effect on the scaffold mechanical properties as well as cell orientation and alignment. MEW scaffolds were mechanically characterized through uniaxial strain testing to determine critical parameters, including strain at failure (SAF), ultimate tensile strength (UTS), Young's modulus (E), fatigue rate, recovery time, and yield strain. These mechanical properties were analysed to define an optimal strain regime for transitioning from static to dynamic culture conditions under muscle-like cyclic loading, relevant to muscle’s viscoelastic behaviour. In parallel, static cultures of human skeletal myotubes and normal human dermal fibroblasts were grown on MEW scaffolds, with varying architectures, to study the effects of fibre aspect ratio on cell alignment. Cell alignment was visualized using DAPI/phalloidin staining and quantified with the ImageJ directionality plugin, enabling a systematic comparison of scaffold designs. This approach evaluates the potential of supportive scaffold architectures to promote aligned cell growth, offering insights into designing effective scaffolds for tissue regeneration.


Validation of the exosomal protein SERPINA11 as a potential atherosclerosis marker via bioprinted scaffold

May 2025

·

1 Read

Existing animal and human cell models have limitations in terms of heterogeneous differences or difficulties in sufficiently reproducing arterial structures and complex cell–cell interactions. The discovery of exosome-derived biomarkers using a 3D bioprinted atherosclerosis model provides a noninvasive and stable detection method and is expected to contribute to the development of early diagnosis and personalized treatment. To contribute to the discovery of exosome-derived biomarkers related to the early diagnosis and prognosis of cardiovascular diseases using a 3D bioprinted atherosclerosis model, we reproduced an arterial environment using 3D bioprinting composed of a biocompatible extracellular matrix (bioink) and various human cells in vitro. The 3D bioprintedatherosclerosis model composed of inflammatory macrophages, coronary artery smooth muscle cells, coronary artery endothelial cells, and collagen methacryloyl (ColMA) hydrogel was treated with LDL to induce atherosclerosis, and the atherosclerosis model was classified into Baseline (BL), Early Atherosclerosis (EA; Early Athero), and Late Atherosclerosis (LA; Late Athero) groups. The secreted exosomes were isolated according to the time period, and a characterization analysis was conducted to confirm the purity of the isolated exosomes. We evaluated the isolated exosomes qualitatively and quantitatively. Isolated exosomes were analyzed using proteomics and miRNA sequencing to verify whether the bioprinted atherosclerosis model induced atherosclerosis, and a novel early atherosclerosis biomarker, SERPINA11, was discovered. In conclusion, we verified that the bioprinted atherosclerosis model induced atherosclerosis and that the novel biomarker set of exosomal miRNAs (hsa-miR-143-5p and hsa-miR-6879-5p) expressed in early atherosclerosis and proteins (SERPINA11, AHSG, and F2) might be clinically useful in early diagnosis and prognosis.


This schematic diagram illustrates the anatomy of the human ear and the biological background of hearing loss. (a) Overview of causes of hearing loss (conductive and sensorineural hearing loss). (b) Detailed anatomical structure of the human ear, divided into the outer, middle, and inner ear, with labeled components. (c) Mechanisms of conductive hearing loss that can lead to mucus buildup, Eustachian tube blockage, and eardrum perforation, causing impaired sound conduction. (d) Sensorineural hearing loss mechanisms depicting the cochlear structure and damage affecting the auditory signal transmission to the brain.
The evolution of bioengineering assisted hearing research showcases groundbreaking milestones and pioneering advancements that have revolutionized treatments for both conductive and sensorineural hearing loss.
Significant impact of tissue engineering on restoring hearing loss. (a) Schematics of micro/nano-scale platforms assisted ear tissue engineering approaches for hearing recovery; (b) fabrication of patient specific PCL based ear shaped cartilage using 3D-printing for auricular reconstruction. Reprinted from [114], Copyright (2018), with permission from Elsevier; (c) radially aligned electro-spun nanofibrous patches incorporated with epidermal growth factor for chronic tympanic membrane perforations. [151] John Wiley & Sons. © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim. Reprinted from [152], Copyright (2016), with permission from Elsevier; (d) fabrication of patient specific PDMS-based 3D printed biomimetic cochlear implants with machine learning. Reproduced from [123]. CC BY 4.0; (e) decellularized cochlea as compatible scaffolds for inner ear engineering. Reproduced with permission from [154]. Reproduced from [124]. CC BY 4.0; (f) generation of mature inner ear organoids with incorporated graphene oxide (GO) for enhanced functionality. Reprinted with permission from [122]. Copyright (2023) American Chemical Society. Reproduced from [112], with permission from Springer Nature.
The role of micro/nanoengineering based enhanced drug delivery approaches on restoring hearing loss. (a) Schematics of micro/nano-scale platforms to significantly enhance the current drug delivery approaches for hearing recovery; (b) microfluidic gelatin methacryloyl microgels based local drug delivery platform for efficient hearing loss therapy. Reprinted with permission from [165]. Copyright (2022) American Chemical Society; (c) a topic drug delivery system using lipid nanoparticles (transfersomes) for efficient delivery through middle and inner ear. Reprinted with permission from [167]. Copyright (2022) American Chemical Society; (d) a novel self-degradable N-hexanoyl glycol chitosan based thermogel with conjugated dexamethasone for enhanced inner ear drug delivery. Reprinted from [168], Copyright (2023), with permission from Elsevier; (e) fabrication of a macro-micro manipulation platform composed of an automated robotics-based approach with a linear actuator to precisely pull and steer magnetic microparticles to ease inner ear drug delivery. Reprinted from [169], Copyright (2018), with permission from Elsevier.
The role of micro/nanoengineering based sensory stimulation approaches on restoring hearing loss. (a) Illustrations of micro/nano-scale platform based compatible electrical and optical stimulation strategies for hearing recovery; (b) electrical stimulation-based approach using ultrafine piezoelectric fiber composites for cochlear stimulation. Reprinted from [181], Copyright (2010), with permission from Elsevier. Reprinted from [138], Copyright (2017), with permission from Elsevier; (c) synthesis of electro-spun piezoelectric polymeric nanofibers based multi-resonant acoustic devices implantable auditory sensors. Reproduced from [182]. CC BY 4.0; (d) development of optical cochlear implants based on optogenetic stimulation. Reproduced from [183]. © The Author(s). Published by IOP Publishing Ltd. CC BY 4.0; (e) optical hearing restoration platform using computational design based human cochlear modeling with light-emitting diode (LED) or laser-coupled waveguide emitters of the optical CI. Reprinted from [26]. © 2022 The Author(s). Published by Elsevier B.V. on behalf of Research Network of Computational and Structural Biotechnology.
Ultra-tiny-scale technology for engineering human ear therapeutics

Ultra-tiny-scale technology representing engineered micro- and nano-scale materials has gained considerable attention for a wide range of applications, including hearing restoration. The advent of hearing loss and its recovery has been the topic of intense discussion since many decades. Although conventional treatments partially support hearing recovery, they present certain limitations such as subsequent immune response and donor site morbidity leading to even worsened sensory disturbances. Microscale- and nanoscale-based approaches such as tissue engineering, nanoparticle-assisted drug delivery systems, and micro/nanofabrication-aided auditory stimulations have been shown to play an efficient role in recovery from hearing disorders. In particular, the introduction of different biomaterials and biopolymers (natural and synthetic) with influential topographical cues and excellent biocompatibility has been found to conveniently bypass previous challenges posed by rigid human ear structures and provided a new path for improved and advanced hearing-recovery approaches. This review is focused on the development of micro/nanoengineering-based hearing recovery therapeutics and their significant impact on the future of hearing research. It discusses the physiological functions associated with the human ear and the mechanism underlying distinct hearing loss disorders as well as highlights various engineered ultra-tiny-scale-assisted strategies for developing advanced hearing therapeutics. Finally, we deliberate on commercialization aspect and future perspectives of implementing micro/nanotechnologies for hearing restoration platforms.


Bioprinted M2 macrophage-derived extracellular vesicle mimics attenuate foreign body reaction and enhance vascularized tissue regeneration

Foreign body reaction (FBR) and insufficient vascularization greatly hinder the integration of 3D-bioprinted tissue substitutes with host tissues. Previous studies have shown that these problems are exacerbated by the stiffness of the 3D-bioprinted constructions, which is highly associated with the abnormal polarization of macrophages. Therefore, we developed an engineering strategy using membrane extrusion to prepare macrophage-derived extracellular vesicle mimics (EVMs). The EVMs derived from M1 and M2 macrophages (M1-EVMs and M2-EVMs) were rich in functional proteins. In the 2D environment, M1-EVMs promoted the fibrotic phenotype of fibroblasts, vascularization, and the M1 polarization of macrophages. In contrast, M2-EVMs effectively avoided the fibrotic trend, showed stronger angiogenic capabilities, and prevented excessive M1 polarization, demonstrating their potential to inhibit FBR and promote neovascularization. After bioprinting the EVMs loaded by gelatin-alginate bioink, the basic physical properties of the bioink were not significantly affected, and the biological functions of EVMs remain stable, indicating their potential as bioink additives. In the subcutaneous implantation model, unlike the FBR-aggravating effects of M1-EVMs, 3D-bioprinted M2-EVMs successfully reduced the immune response, prevented fibrous capsule formation, and increased vascular density. When applied to skin wound treatment, 3D-bioprinted M2-EVMs not only inhibited inflammatory levels but also exhibited pleiotropic pro-regenerative effects, effectively promoting vascularization, re-epithelialization, and appendage regeneration. As an innovative additive for bioinks, M2-EVMs present a promising approach to enhance the survival of bioengineered tissues and can further serve as a targeted drug loading system, promoting the development of regenerative medicine and improving clinical outcomes.


Chip design considerations and printing quality. (a) Schematic of molding approach. (b) Schematic illustration and fluorescent image of issue with cells floating out of the chip, represented by fluorescent beads. Scale bar = 2 mm. (c) Schematic and bright-field (BF) image of air bubbles (red arrows) arising from inserting plug. Scale bar = 1 mm. (d) Schematic and fluorescent image of U-shaped chip designs which allows for more reliable cell seeding here illustrated by the injection of fluorescent beads. Scale bar = 2 mm. (e) Percentage deviations of the printed chip from original CAD model. (f) Predicted light intensity normalized to the median light intensity over the whole 3D model. (g) Percentage deviation of printed chip zoom in showing narrowing of the channel at the inlets. (h) Cross section of predicted light intensity at the channel inlet. (i)–(k) Printing accuracy for X, Y and Z coordinates.
Seeding setup and optimization. (a) Schematic of rotation setup. (b) Fluorescent images of chips seeded with (left) and without (right) rotation. (c) Percentage of the channel surface covered by cells with and without rotation. (d) Channels seeded with MCF10A with different coatings. (e) Percentage of the channels (length = 9 mm diameter = 1 mm) covered with cells with different cell coatings. (f) Channels seeded with different densities of MCF10A and (g) percentage channel coverage. (h) Channels seeded with HUVECs at 20 M cells ml⁻¹ with different coatings. (i) Channel coverage of HUVECs with different coatings. Scale bars = 100 µm. Images were taken one week after cell seeding. Differences in cell coverage according to One-way ANOVA were not significant due to sample size unless indicated otherwise (* for p < 0.05).
Bioreactor setup. (a) Schematic of bioreactor parts and functions. (b) 3D printed bioreactor. (c) CNC machined bioreactor. (d) Perfusion setup. Zoom in: Tubing clips.
Influence of perfusion on cellular organization of mammary epithelial cells. (a) Percentage expansion patterns of channels under perfusion. (b) Plot of the channel edges at maximum and minimum distance. (c) BF images of perfused and non-perfused channels showing cell invasion into the surrounding tissue for perfused chips as assessed through measurements in Fiji. Scale bar = 500 µm. (d) FITC-Inulin perfusion into seeded channels and FITC quantification outside the channel boundaries over time, as well as (e) quantification of the area under the curve outside of the channel boundaries, demonstrating epithelial barrier function under perfusion. Scale bar = 500 µm. (f) Immunofluorescent images of non- perfused and perfused chips (g). Scale bar = 100 µm. (h) Percentage of cells lining the inner edge of the duct that are K8/18 positive. (i) Percentage of cells lining the duct for which the inner layer is K8/18 positive and is followed by K14 positive cells. Data was acquired after two weeks of perfusion. Differences between culture conditions were not statistically significant due to sample size unless indicated otherwise (* for p < 0.05) as calculated through One-way ANOVA (e) and unpaired t-test (h), (i).
Strategies to improve model maturation. (a) Fluorescent images of chips seeded with MCF10A or HUVECs displaying cellular alignment along the channels circumference. Scale bar = 100 µm (b) Schematic and BF images of chip printed in vertical orientation. Scale bar = 250 µm. (c) Schematic and BF images of chip printed in horizontal orientation. Scale bar = 250 µm. (d) Fluorescent images of horizontally printed chips seeded with MCF10A or HUVECs displaying longitudinal cell alignment. Scale bar = 100 µm. (e) Quantification of cell alignment by print orientation. (f) Nominal comparison of horizontally printed chip indicating reduced channel diameters along longitudinal axis. (g) Computed light intensities over the model when printed horizontally indicating high light accumulation within horizontally printed part of the duct. (h) Schematic, BF images (i) and fluorescent images (j) of chip containing an epithelial duct with lobular structures. Scale bar = 1 mm. (k) Fluorescent image showing MCF10A invasion from the lobular structures (k). Scale bar = 200 µm (k) scale bar = 100 µm (l) (m) schematic of chip with two channels of 200 µm distance. Scale bar = 200 µm. (n) Fluorescent images of 2-channel chip demonstrating the separate culture of these two cell types. Scale bar = 200 µm. Statistical significance of differences between vertically and horizontally printed chips was assessed using Welch’s t-test (** for p < 0.01; * for p < 0.05).
Development of a bioreactor and volumetric bioprinting protocol to enable perfused culture of biofabricated human epithelial mammary ducts and endothelial constructs

Tissue function depends on the 3D spatial organization of cells, extracellular matrix components, as well as dynamic nutrient gradients and mechanical forces. Advances in biofabrication technologies have enabled the creation of increasingly sophisticated tissue models, but achieving native-like tissue maturation post-fabrication remains a challenge. The development of bioreactors and microfluidic systems capable of introducing dynamic culture platforms and controlled mechanical and biochemical stimulation for biofabricated tissue analogues is therefore imperative to address this. In this technical note, we introduce a multi-step pipeline to fabricate, seed and perfuse geometrically complex hydrogel constructs with quality control protocols through the computational analysis of confocal multispectral 3D imaging data for each step of the process. Employing ultra-fast volumetric bioprinting, chips with tunable channel architectures were fabricated. Furthermore, an autoclavable and transparent perfusion bioreactor inspired by open-source designs was developed to enable controlled, long-term perfusion (up to 28 days) and real-time monitoring of cell behavior. As proof-of-concept, employing this pipeline, we fabricated a human mammary ductal model and an endothelialized vessel on-a-chip, demonstrating the compatibility of the platform with epithelial and endothelial cell lines, and investigated the effect of dynamic culture on tissue-specific cell organization. Dynamic perfusion underlined the influence of mechanical stimulation on cell organization and maturation. Various chip architectures, capable of recapitulating tissue-specific features (i.e. lobules) were printed, enabling the mono- and co-culture of human mammary epithelial and endothelial cells. Our pipeline, with the accompanying protocols and analysis scripts presented here, provide the potential to be applied for the dynamic culture of a wide range of tissues.


Development of small tissue engineered blood vessels and their clinical and research applications

May 2025

·

10 Reads

Since the first tissue engineered blood vessel (TEBV) was developed, different approaches, biomaterial scaffolds and cell sources have been used to obtain an engineered vessel as much similar as native vessels in terms of structure, functionality and mechanical properties. At the same time, diverse needs to obtain a functional TEBV have emerged, such as for blood vessel replacement for cardiovascular diseases to be used as artery bypass, to vascularize tissue engineered constructs, or even to model vascular diseases or drug testing. In this review, after briefly describing the native structure and function of arteries, we will give an overview of different biomaterials, cells and methods that have been used during the last years for the development of small TEBVs (1-6 mm diameter). The importance of perfusing the TEBVs in order to acquire functionality and maturation will be also discussed. Finally, we will center the review on TEBV applications beyond their use as vascular graft for cardiovascular diseases.


3D bioprinting of human iPSC-derived cardiac constructs with microvascular network support for improved graft survival in vivo

May 2025

·

41 Reads

Cardiac tissue engineering is a rapidly growing field that holds great promise for the development of new therapies for heart disease. While significant progress has been made in the field over the past two decades, engineering functional myocardium of clinically relevant size and thickness remains an unmet challenge. A major roadblock in this respect is the current difficulty in incorporating efficient vascularization into engineered constructs. One potential solution involves the use of microvascular fragments from adipose tissue, which have demonstrated encouraging results in improving vascularization and graft survival following transplantation. However, this method lacks precise control over the vascular architecture within the constructs. Here, we set out to investigate the use of 3D bioprinting for the fabrication of human cardiac tissue constructs composed of human induced pluripotent stem cell (hiPSC) derivatives, while allowing for the precise control of the distribution and density of microvessel fragments within the bioprinted constructs. We carefully selected and optimized bioink compositions based on their printability, biocompatibility, and construct stability. Following transplantation into immunodeficient mice, 3D bioprinted cardiac constructs containing microvessel fragments exhibited rapid and efficient vascularization, resulting in prolonged graft survival. Overall, our studies underscore the advantages of employing engineering design and self-assembly across different scales to address current limitations of tissue engineering, and highlight the usefulness of 3D bioprinting in this context.


Biofabrication of microstructured bacterial ecosystems using chaotic bioprinting: advancing in vitro research for microbial engineering

May 2025

·

7 Reads

Mixed microbial communities are essential for various ecosystems, with bacteria often exhibiting unique behaviors in structured environments. However, replicating these interactions in vitro remains challenging, as traditional microbiology techniques based on well-mixed cultures fail to capture the spatial organization of natural communities. Chaotic 3D printing offers a versatile, high-throughput method for fabricating hydrogel constructs with multilayered microstructure in which different bacterial strains can coexist, closely mimicking the partial segregation seen in natural microbial ecosystems. Using a Kenics static mixer (KSM) printing nozzle, we bioprinted a bacterial consortium consisting of Lactobacillus rhamnosus, Bifidobacterium bifidum, and Escherichia coli as a simplified model for human gut microbiota. Chaotic bioprinting enabled the creation of microstructured cocultures with distinct niches, allowing all bacterial strains to coexist (without being scrambled) and reach a population equilibrium. We characterized the cocultures through fluorescence microscopy, colony counting, and quantitative polymerase chain reactions (qPCR). Our results demonstrate that the microarchitecture of the printed fibers significantly influences bacterial growth dynamics. Stratified arrangements enhanced coculture viability and balance over 72 hours compared to well-mixed and suspension conditions. Chaotic printing also allows the rational arrangement of strict anaerobic bacteria, such as B. bifidum, by positioning them in construct layers that are more susceptible to hypoxia. Chaotic bioprinting presents a powerful tool for engineering microbial ecosystems with precise spatial control. This approach holds promise for advancing our understanding of microbial interactions and has potential biomedical applications in antibiotic testing, microbiota research, bioremediation, and synthetic biology.


Acoustic holographic assembly of cell-dense tissue constructs

May 2025

·

7 Reads

Tissue engineering aims to develop tissue constructs as models or substitutes for native tissues. For organ-level biological studies and regenerative medicine applications, it is essential to fabricate tissue constructs with physiologically relevant cell densities (on the order of 10 million to 1 billion cells·mL ⁻¹ , large size (centimeter scale and larger), and a controllable geometry to guide tissue maturation. State-of-the-art biofabrication methods, however, struggle to simultaneously meet all of these demands. The recently proposed acoustic holographic assembly (AHA) method shows promise, as it is compatible with culture media and enables the contactless, label-free, and volumetric assembly of biological cells in a predefined geometry within few minutes. Here we present an AHA biofabrication scheme designated for fabricating cell-dense, centimeter-scale, and arbitrarily-shaped tissue constructs using a compact benchtop instrument compatible with a biolab environment. We demonstrate the assembly of C2C12 myoblasts in gelatin methacryloyl (GelMA) into large and asymmetric branch-shaped constructs, which are rapidly formed with an average cell density of 40 million cells·mL ⁻¹ and a local density of up to 260 million cells·mL ⁻¹ . Featuring a high viability of 90.5%±4.3%, the assembled cell constructs are observed to grow within the GelMA hydrogel under perfusion over five days. Further, we show how AHA can --- in a single step --- assemble cells into layered and three-dimensional geometries inside standard cell culture labware. It can therefore help obtain engineered tissue constructs with structural and functional characteristics seen in more complex native tissues.


Controlled microvasculature for organ-on-a-chip applications produced by high-definition laser patterning

May 2025

·

19 Reads

Organs-on-Chips (OoCs) are 3D models aiming to faithfully replicate in vitro specific functions of human organs or tissues. While promising as an alternative to traditional 2D cell culture and animal models in drug development, controlled realization of complex microvasculature within OoC remains a significant challenge. Here, we demonstrate how femtosecond laser patterning allows to produce hollow microvascular-like channels inside a collagen-based matrix directly within a microfluidic chip. The hydrogel preparation protocol was optimized to maintain structural stability, facilitating successful endothelialization of produced channels. The resulting microvascular structures exhibit notable physiological relevance, as evidenced by the expression of key endothelial markers (ZO-1, and VE-cadherin) and the successful reproduction of the barrier function. Furthermore, tumor necrosis factor-alpha (TNF-α) exposure induces a concentration-dependent increase in vascular permeability and inflammatory marker expression (ICAM-1). The proposed method holds the potential to control and faithfully reproduce the vascularization process in OoC platforms, in both physiological and inflammatory conditions.


Nuclei morphologies in healthy and unhealthy human tendons. (a) Photographs of healthy and unhealthy human tendons alongside representative confocal microscopy images showing cell nuclei and collagen type I. The right column depicts the orientation color map obtained from the OrientationJ plugin, corresponding to the confocal microscopy images. Scale bars: 20 µm. (b) Nuclei aspect ratio (nAR) of cells and its distribution of nAR in healthy and unhealthy tendons (6 patients, dataset size > 125). (c) Cellularity comparison between healthy and unhealthy tendons. Additional details about the patients are available in the supporting information. (d) Percentage of aligned collagen fibers in healthy and unhealthy human tendons. Collagen fibers are classified as aligned, characterized by an angle deviation from the peak distributed direction between −10 to 10°.
Schematic illustration of study design: This figure depicts the working principle of filamented light (FLight) biofabrication of the tissue-engineered tendon constructs (mini-tendon). Freshly isolated human tenocytes are expanded in 2D culture and mixed with Gel-MA photoresin to prepare the bioresin. Due to the optical modulation instability (OMI), the speckle pattern, characterized by local maxima and minima intensity distributions, is present in the projected laser beams, leading to filamentation of light. Through modulation by a digital micromirror device (DMD), pre-designed geometric images are projected onto a resin pool. When the filamented light illuminates to an optically non-linear medium (i.e. (bio)resin), a self-focusing effect occurs, guiding the filamented light to photo-crosslink the (bio)resin into microfilaments. After the removal of uncrosslinked (bio)resin, microchannels are formed within the hydrogel constructs. These hydrogels, featuring various microfilaments and microchannels, create different matrix microenvironments (i.e. mechanical confinements) for the encapsulated tenocytes, leading to distinct cell/nuclei morphologies and diverse cellular behaviors. Up to 96 in vitro hydrogel constructs in millimeter scale are created within a matter of seconds. These constructs are then transferred to culture plates for subsequent incubation and analysis. Graphics were created in part using BioRender. Created in BioRender. Zenobi-wong, M. (2025) https://BioRender.com/mo901do.
Characterization of microarchitectures in varying FLight hydrogel matrices. (a) Representative confocal microscopy images showcasing microfilaments and microchannels across different hydrogel matrices (Matrix 1–4). The top row presents a 3D view of microarchitectures, generated from 3D reconstruction of Z-stack scans using Imaris 9.9.0 software. The middle row displays maximum-intensity projection images from Z-stack scans. The bottom row features an orientation color map obtained from the OrientationJ plugin, corresponding to the projection images. Scale bars: 50 µm. (b) Distribution of microfilament orientation in various hydrogel matrices (Matrix 1–4). Gray areas highlight microfilaments classified as aligned, characterized by an angle deviation from the projection direction between −10 to 10°. Numeric labels represent the percentage of aligned microfilaments. (c) Mechanical properties of hydrogel matrices evaluated using compression modulus (n = 4). The compressions were applied in parallel to the microfilament direction. (d) Analysis of microfilament and microchannel diameters across different FLight matrices (n = 3, dataset size = 50). The above values indicate the mean diameters. (e) The ratio of microchannels in the total 3D hydrogel volume, indicating the porosity of the FLight matrices (n = 3, dataset size > 18).
Nuclei morphologies in mini-tendon constructs. (a) Representative confocal microscopy images showing human tenocytes in different mini-tendon constructs after 0, 3, 7, and 14 d of culture. The column labels indicate the specific matrices (as depicted in figure 3) in which the cells were encapsulated. Images are maximum-intensity projections, scale bar: 50 µm. (b) Analysis of nuclei aspect ratio (nAR) in human tenocytes at different culture durations, and c) the distribution of nAR in Matrix 1–4 following 14 d of culture (n = 3, dataset size > 22). (d) Cellularity comparison within Matrix 1–4 at day 0, 3, 7, and 14. (e) Proportion of aligned human tenocytes in different mini-tendon constructs, measured after 7 d (left) and 14 d (right) of incubation. The aligned cells are characterized by an angle deviation of f-actin from the projection direction between −10 to 10° using F-actin staining.
Nuclei morphologies and YAP nuclear activation of tenocytes under different mechanical confinements. (a) Representative confocal microscopy images of human tenocytes within various mini-tendon constructs (Matrix 1 and 4), and on the 2D surface of FLight hydrogel constructs (Matrix 1 and 4) after 14 d of culture. Scale bar: 50 µm. (b) Quantification of aligned f-actin in human tenocytes. F-actin alignment is defined by an angle deviation from the peak distributed direction between −10 to 10°. (c) Analysis of the nuclei aspect ratio (nAR) in or on different hydrogel matrices, along with the distribution of nAR following 14 d of culture (n = 3, dataset size > 109). (d) Representative images showing the nuclear morphologies of tenocytes and the YAP intensity distribution map. The yellow dashed line indicates the nuclei location. The 3D reconstruction of the nuclei was performed using Imaris 9.9.0 software, with relative intensity displayed on a rainbow scale. (e) Measurement of nuclear volume and f) the overlap ratio of nuclei to YAP. A ratio of 0 indicates an absence of YAP signal within the nuclei, whereas a ratio of 1 denotes the presence of YAP signals encompassing the entire nuclear volume (n = 3, dataset size > 109).
Filamented light (FLight) biofabrication of mini-tendon models show tunable matrix confinement and nuclear morphology

April 2025

·

10 Reads

·

1 Citation

One hallmark of healthy tendon tissue is the high confinement of tenocytes between tightly packed, highly aligned collagen fibers. During tendinopathy, this organization becomes dysregulated, leading to cells with round-shaped morphology and collagen fibers which exhibit crimping and misalignment. The elongated nuclei in healthy tendons are linked to matrix homeostasis through distinct mechanotransduction pathways, and it is believed that the loss of nuclear confinement could upregulate genes associated with abnormal matrix remodeling. Replicating the cell and nuclear morphology of healthy and diseased states of tendon, however, remains a significant challenge for engineered in vitro tendon models. Here we report on a high throughput biofabrication of mini-tendons that mimick the tendon core compartment based on the filamented light (FLight) approach. Each mini-tendon, with a length of 4 mm, was composed of parallel hydrogel microfilaments (2–5 µm diameter) and microchannels (2–10 µm diameter) that confined the cells. We generated four distinct matrices with varying stiffness (7–40 kPa) and microchannel dimensions. After 14 d of culture, 29% of tenocytes in the softest matrix with the largest microchannel diameter were aligned, exhibiting an average nuclear aspect ratio (nAR) of 2.1. In contrast, 84% of tenocytes in the stiffest matrix with the smallest microchannel diameter were highly aligned, with a mean nAR of 3.4. When tenocytes were cultured on the FLight hydrogels (2D) as opposed to within the hydrogels three-dimensional (3D), the mean nAR was less than 1.9, indicating that nuclear morphology is significantly more confined in 3D environments. By tuning the stiffness and microarchitecture of the FLight matrix, we demonstrated that mechanical confinement can be modulated to exert control over the extent of nuclear confinement. This high-throughput, tunable platform offers a promising approach for studying the mechanobiology of healthy and diseased tendons and for eventual testing of drug compounds against tendinopathy.


BigMACS geometry and FEA mesh sensitivity analysis. (A) Illustration of the BigMACS platform, showing the presence of microgrooves in the cell culture chamber, with confocal imagery of the 22 µm tall groove height. (B) Mechanical stress results of micro-grooved BigMACS under radial distension with various FEA mesh resolutions, showing inconsistent stress profiles below 10 000 mesh nodes per mm². (D) Sensitivity of FEA mesh node density on Von Mises stress at top, bottom, and mid-point slope areas. Subfigure A is a modified version of supplemental data from [18]. Adapted from [18]. © IOP Publishing Ltd. All rights reserved.
Biomechanics simulations of BigMACS. Full-scale strain and stress-magnitude simulations of BigMACS under radial distension, angular flexion, and radial and angular distension. (Right) Stress magnitude (3D Von Mises Stress) and stress direction (XY Mohr’s Angle) of a cell culture region imaged by confocal microscopy.
Quantitative histology of BigMACS conditioning. (A) MSCs were cultured in soft robotic bioreactors for four days in static (left), or for three days in static and one day in radial, angular, or radial and angular mechanical conditioning (right) and imaged by 4 × 2 tiled 3D confocal microscopy. Scale bars are 0.05 cm. (B) Computational histology analyses of the nuclear density, actin orientation, α-SMA stain density, and Col-IV stain density of the four BigMACS conditions (note that heatmap scales for the static control condition are several-fold lower than mechanically-active conditions).
Impact of 3D Von Mises stress on BigMACS cell behaviours. Scatterplots of 3D Von Mises stress (kPa; x-axis) versus nuclear (DAPI stain) density, α-SMA stain density, and Col-IV stain density per cm² or average actin orientation, as calculated in a 50 µm radius around each detected stain. Third-order polynomials were fitted to the imaging data with 95% confidence intervals calculated. Regression statistical replicates were calculated as the number of stains detected per image (1201–451 749) and not imaging, experimental, or biological replicates.
Impact of XY Mohr’s stress angle on BigMACS cell behaviours. Scatterplots of XY Mohr’s stress angle (x-axis) versus nuclear (DAPI stain) density, α-SMA stain density, and Col-IV stain density per cm² or average actin orientation, as calculated in a 50 µm radius around each detected stain. Third-order polynomials were fitted to the imaging data with 95% confidence intervals calculated. Regression statistical replicates were calculated as the number of stains detected per image (1201–451 749) and not imaging, experimental, or biological replicates.
Simulating big mechanically-active culture systems (BigMACS) using paired biomechanics-histology FEA modelling to derive mechanobiology design relationships

April 2025

·

16 Reads

Big mechanically-active culture systems (BigMACS) are promising to stimulate, control, and pattern cell and tissue behaviours with less soluble factor requirements. However, it remains challenging to predict if and how distributed mechanical forces impact single-cell behaviours to pattern tissue. In this study, we introduce a tissue-scale finite element analysis framework able to correlate sub-cellular quantitative histology with centimetre-scale biomechanics. Our framework is relevant to diverse BigMACS, including media perfusion, tensile-stress, magnetic, and pneumatic tissue culture platforms. We apply our framework to understand how the design and operation of a multi-axial soft robotic bioreactor can spatially control mesenchymal stem cell (MSC) proliferation, orientation, differentiation to smooth muscle, and extracellular vascular matrix deposition. We find MSC proliferation and matrix deposition to positively correlate with mechanical stimulation but cannot be locally patterned by soft robot mechanical stimulation within a centimetre scale tissue. In contrast, local stress distribution was able to locally pattern MSC orientation and differentiation to smooth muscle phenotypes, where MSCs aligned perpendicular to principal stress direction and expressed increased α-SMA with increasing 3D Von Mises Stresses from 0 to 15 kPa. Altogether, our new biomechanical-histological simulation framework is a promising technique to derive the future mechanical design equations to control cell behaviours and engineer patterned tissue.


Cardiomyocyte sheet stacking using fibrin enables high-speed construction of three-dimensional myocardial tissue and high transplantation efficiency

April 2025

·

30 Reads

Despite the development of three-dimensional (3D) tissues that promises remarkable advances in myocardial therapies and pharmaceutical research, vascularization is required for the repair of damaged hearts using cardiac tissue engineering. In this study, we developed a method for rapid generation of a 3D cardiac tissue, with extremely high engraftment efficiency, by stacking cardiomyocyte sheets using fibrin as an adhesive. Cell sheets were created by peeling off confluent cultured cells from a culture dish grafted with a polymer that induced surface hydrophilicity in response to low temperatures. The high engraftment rate was attributed to the retention of the adhesive protein. The multistacked vascularized cell sheets prepared using fibrin, when transplanted into the subcutaneous tissue and at myocardial infarction site in rats, yielded a transplanted 3D myocardial tissue. Furthermore, multilayered cardiomyocyte sheets were transplanted twice at 1 week intervals to create a 3D myocardial tissue. Our data suggest that fibrin-based rapidly layered cell sheets can advance tissue-engineered transplantation therapy and should aid the development of next-generation tissue-engineered products in the fields of regenerative medicine and drug screening.


Injectable microgel and micro-granular hydrogels for bone tissue engineering

Injectable microgels, made from both natural and synthetic materials, are promising platforms for the encapsulation of cells or bioactive agents, such as drugs and growth factors, for delivery to injury sites. They can also serve as effective micro-scaffolds in bone tissue engineering (BTE), offering a supportive environment for cell proliferation or differentiation into osteoblasts. Microgels can be injected in the injury sites individually or in the form of aggregated/jammed ones named micro-granular hydrogels. This review focuses on common materials and fabrication techniques for preparing injectable microgels, as well as their characteristics and applications in BTE. These applications include their use as cell carriers, delivery systems for bioactive molecules, micro-granular hydrogels, bio-inks for bioprinting, three-dimensional microarrays, and the formation of microtissues. Furthermore, we discuss the current and potential future applications of microgels in bone tissue regeneration.


Schematic illustrations of (a) microgel generation by a flow-focusing microfluidic device and different microgel compositions for (b) bone and (c) cartilage microtissues.
Combinatorial strategy for engineering cartilage and bone microtissues using microfluidic cell-laden microgels

Osteochondral defects (OCD) refer to localized injuries affecting both the avascular cartilage and subchondral bone. Current treatments, such as transplantation or microfracture surgery, are hindered by limitations like donor availability and the formation of small, rigid fibrocartilage. Tissue engineering presents a promising alternative, yet challenges arise from limited oxygen and nutrient supply when fabricating human-scale tissue constructs. To address this, we propose assembling engineered micro-scale tissue constructs as building blocks for human-scale constructs. In this study, we aimed to develop bone and cartilage microtissues as building blocks for osteochondral tissue engineering. We fabricated placental stem cell (PSC)-laden microgels, inducing differentiation into osteogenic and chondrogenic microtissues. Utilizing a microfluidics chip platform, these microgels comprised a cell-laden core containing bone-specific and cartilage-specific growth factor-mimetic peptides, respectively, along with an acellular hydrogel shell. Additionally, we investigated the effect of culture conditions on microtissue formation, testing dynamic and static conditions. Results revealed over 85% cell viability within the microgels over 7 d of continuous growth. Under static conditions, approximately 60% of cells migrated from the core to the periphery, while dynamic conditions exhibited evenly distributed cells. Within 4 weeks of differentiation, growth factor-mimetic peptides accelerated PSC differentiation into bone and cartilage microtissues. These findings suggest the potential clinical applicability of our approach in treating OCD.


Review of 3D-printed bioceramic/biopolymer composites for bone regeneration: fabrication methods, technologies and functionalized applications

Biomaterials for orthopedic applications must have biocompatibility, bioactivity, and optimal mechanical performance. A suitable biomaterial formulation is critical for creating desired devices. Bioceramics with biopolymer composites and biomimetics with components similar to that of bone tissue, have been recognized as an area of research for orthopedic applications. The combination of bioceramics with biopolymers has the advantage of satisfying the need for robust mechanical support and extracellular matrices at the same time. Three-dimensional (3D) printing is a powerful method for restoring large bone defects and skeletal abnormalities owing to the favorable merits of preparing large, porous, patient-specific, and other intricate architectures. Bioceramic/biopolymer composites produced using 3D printing technology have several advantages, including desirable optimal architecture, enhanced tissue mimicry, and improved biological and physical properties. This review describes various 3D printing bioceramic/biopolymer composites for orthopedic applications. We hope that these technologies will inspire the future design and fabrication of 3D printing bioceramic/biopolymer composites for clinical and commercial applications.


Biofabrication of small-diameter vascular graft with acellular human amniotic membrane: a proof-of-concept study in pig

Expanded polytetrafluoroethylene (ePTFE) grafts are Food and Drug Administration approved and effective for large vessel surgeries but face challenges in smaller vessels (Inner Diameter, ID ⩽ 6 mm) due to reduced blood flow and higher risks of thrombosis, stenosis, and infection. This study developed a vascular graft with an ID of 6 mm from decellularized human amniotic membrane (DAM graft) and compared its performance to ePTFE grafts in a porcine carotid artery model for one month. DAM grafts retained key extracellular matrix structures and mechanical properties post-decellularization, with customizable layers and stiffness to meet specific clinical needs. DAM grafts demonstrated successful carotid artery replacement, showing good surgical feasibility, patency, and post-operative recovery in all animals. In contrast to ePTFE grafts, which exhibited significant neointimal hyperplasia (NIH), poor endothelialization, and inflammation, DAM grafts displayed organized endothelial coverage, smooth muscle alignment, and reduced inflammation, minimizing NIH, thrombosis, and graft failure. These findings position DAM grafts as a promising alternative to synthetic grafts, especially for small-diameter applications. Future research should focus on improving endothelialization, exploring molecular mechanisms, and assessing long-term outcomes to further optimize DAM grafts for clinical use.


Construction of highly vascularized hepatic spheroids of primary hepatocytes via pro-angiogenic strategy in vitro

April 2025

·

39 Reads

Primary hepatocytes are widely recognized for their ability to accurately represent the in vivo hepatocyte phenotype. However, traditional avascular primary hepatocyte culture models are limited by inadequate mass transfer, which leads to a rapid decline in hepatocyte function and survival. To address these challenges, vascularization of hepatic spheroids is crucial for enhancing oxygen and nutrient supply, thereby enabling the construction of larger and more complex hepatic tissues in vitro. In this study, we achieved vascularization of hepatic spheroids containing freshly isolated primary hepatocytes by incorporating fibroblasts as a source of paracrine factors to induce angiogenesis. Multicellular spheroids composed of primary hepatocytes and fibroblasts were formed in non-adhesive concave wells, and one of the spheroids was subsequently embedded in a fibrin–collagen hydrogel within a microfluidic device. Endothelial cells were then seeded onto adjacent microfluidic channels. They formed microvascular networks that extended toward and penetrated the hepatic spheroid. The vascularized hepatic spheroid closely mimicked hepatic sinusoids, with hepatocytes in close contact with microvessels. Moreover, the vascularized spheroid exhibited significantly enhanced hepatic function, specifically albumin secretion and urea synthesis. Our findings provide insights into the establishment of highly vascularized hepatic spheroids in vitro, which is crucial for constructing scalable hepatic tissues in the context of biofabrication.


BTES for metal additive manufacturing. (a) Porous structure of Ti6Al4V prepared by SLM. Reproduced from Liao et al (2021), with permission from Springer Nature. (b) Porous structure of Ti–Nb alloy prepared by SLM. Reprinted from Luo et al (2020) Copyright (2021), with permission from Elsevier. (c) Ti-6Al-4V scaffold prepared by EBM. Reproduced from Sichani et al (2024). CC BY 4.0.
Laser based powder bed melting process.
Powder particle size distribution of elemental powder mixtures (Ti-35Nb), elemental Nb and CP-Ti powders, morphology of powder mixtures, and EDS analysis of chemical composition of powder mixtures (Ti-35Nb). Reprinted from Wang et al (2021a), Copyright (2021), with permission from Elsevier.
TPMS structure and SEM of butterfly wing and weevil exoskeleton. Han and Che (2018) John Wiley & Sons. © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.
Triply periodic minimal surfaces. Reproduced from Feng et al (2021). CC BY 4.0.
Advancements in selective laser melting (SLM) of titanium alloy scaffolds for bone tissue engineering

April 2025

·

28 Reads

Selective Laser Melting (SLM) has emerged as a transformative technology in bone tissue engineering, particularly for fabricating porous scaffolds from titanium alloys. These scaffolds offer a promising solution for treating critical-sized bone defects, providing mechanical support while promoting bone regeneration. A comprehensive review on recent advancements of SLM is provided by presenting a detailed analysis of cutting-edge research in the application of SLM for titanium alloy scaffold production. Key areas explored include structural designs like Triply Periodic Minimal Surfaces, material and process parameters optimization to enhance scaffold properties such as porosity, mechanical strength, and biocompatibility. Furthermore, the review emphasizes recent innovations in surface modification techniques which improve bioactivity and osseointegration to enable scaffolds to mimic the host tissues. In addition, this review provides essential insights in related to the potential of SLM to be adopted in producing personalized and high-performance medical implants. By synthesizing the latest trends and identifying key areas for future research, this paper aims to serve as a vital resource for the advancement and usage of SLM-fabricated scaffolds in clinical applications. The findings underscore the importance of continued innovation in this field, which has the potential to significantly improve patient outcomes in orthopaedics and beyond.


Fabrication of multilayer heterogeneous cell assembly for pathophysiologically relevant 3D in-vitro IBD disease model for high throughput drug screening

April 2025

·

71 Reads

·

1 Citation

Regarding the approval of novel pharmaceuticals, the most common reason for failure is inadequate oral drug bioavailability. Owing to the complex physiological milieu of the human intestine, which is characterized by its varied composition, various functions, and one-of-a-kind dynamic conditions, it is difficult to reproduce the organ in vitro. Traditional monolayers in two dimensions, sophisticated three-dimensional systems, and developing fluid-dynamic platforms are examples of in-vitro intestinal models. Caco-2 cells have been the gold standard for studying drug permeability for over two decades, particularly for BCS Class II/III/IV drugs. Other intestinal in vitro models exist; however, pharmaceutical corporations and regulatory authorities use the Caco-2 cell line to predict human intestinal permeability. To predict oral drug absorption and study normal intestinal epithelial physiology, it is necessary to have advanced technologies capable of creating human intestinal epithelial cells (hIECs) with cellular variety and functions. There is a strong link between the permeability data obtained in vitro and the fractions absorbed by humans in complex multicellular models. However, although microphysiological systems accurately replicate physiological cues of the digestive tract, they still require standardization. We critically reviewed a step towards tissue-created 3D intestinal organoids and 3D heterocellular multicompartmental models without compromising cellular variety and function. To bridge the gap between 2D and 3D intestinal culture models, a physiologically appropriate hIEC model provides a novel platform for patient-specific testing and translational applications. A comprehensive understanding of numerous 3D in-vitro models of inflammatory bowel disease has been discussed. Additionally, this review will provide insights into the benefits and limitations of these models and their relevance in understanding intestinal physiology and accelerating drug discovery through high-throughput screening.


Journal metrics


9.8 (2023)

Journal Impact Factor™


22%

Acceptance rate


17.4 (2023)

CiteScore™


5 days

Submission to first decision


128 days

Submission to publication


2.5 (2023)

Immediacy Index


1.769 (2023)

SJR


£2,720 / € 3,100 / $3,800

Article processing charge

Editors