bims-biprem Biomed News
on Bioprinting for regenerative medicine
Issue of 2025–01–26
ten papers selected by
Seerat Maqsood, University of Teramo



  1. J Tissue Eng. 2025 Jan-Dec;16:16 20417314241308022
      The field of three dimensional (3D) bioprinting has witnessed significant advancements, with bioinks playing a crucial role in enabling the fabrication of complex tissue constructs. This review explores the innovative bioinks that are currently shaping the future of 3D bioprinting, focusing on their composition, functionality, and potential for tissue engineering, drug delivery, and regenerative medicine. The development of bioinks, incorporating natural and synthetic materials, offers unprecedented opportunities for personalized medicine. However, the rapid technological progress raises regulatory challenges regarding safety, standardization, and long-term biocompatibility. This paper addresses these challenges, examining the current regulatory frameworks and the need for updated guidelines to ensure patient safety and product efficacy. By highlighting both the technological potential and regulatory hurdles, this review offers a comprehensive overview of the future landscape of bioinks in bioprinting, emphasizing the necessity for cross-disciplinary collaboration between scientists, clinicians, and regulatory bodies to achieve successful clinical applications.
    Keywords:  3D bioprinting; bioinks; biomedical application; crosslinking
    DOI:  https://doi.org/10.1177/20417314241308022
  2. Adv Healthc Mater. 2025 Jan 24. e2404925
      Spinal cord injury (SCI) leads to acute tissue damage that disrupts the microenvironmental homeostasis of the spinal cord, inhibiting cell survival and function, and thereby undermining treatment efficacy. Traditional stem cell therapies have limited success in SCI, due to the difficulties in maintaining cell survival and inducing sustained differentiation into neural lineages. A new solution may arise from controlling the fate of stem cells by creating an appropriate mechanical microenvironment. In this study, mechanical response stem cell complex (MRSCC) is created as an innovative therapeutic strategy for SCI, utilizing 3D bioprinting technology and gelatin microcarriers (GM) loaded with mesenchymal stem cells (MSCs). GM creates an optimal microenvironment for MSCs growth and paracrine activity. Meanwhile, 3D bioprinting allows accurate control of spatial pore architecture and mechanical characteristics of the cell construct to encourage neuroregeneration. The mechanical microenvironment created by MRSCC is found to activate the Piezo1 channel and prevent excessive nuclear translocation of YAP, thereby increasing neural-related gene expression in MSCs. Transplanting MRSCC in rats with spinal cord injuries boosts sensory and motor recovery, reduces inflammation, and stimulates the regeneration of neurons and glial cells. The MRSCC offers a new tissue engineering solution that can promote spinal cord repair.
    Keywords:  3D bioprinting; mechanical responses; mesenchymal stem cells; microcarriers; spinal cord injuries
    DOI:  https://doi.org/10.1002/adhm.202404925
  3. J Tissue Eng. 2025 Jan-Dec;16:16 20417314241313341
      The development of advanced in vitro models for assessing liver toxicity and drug responses is crucial for personalized medicine and preclinical drug development. 3D bioprinting technology provides opportunities to create human liver models that are suitable for conducting high-throughput screening for liver toxicity. In this study, we fabricated a humanized liver model using human-induced hepatocytes (hiHeps) derived from human fibroblasts via a rapid and efficient reprogramming process. These hiHeps were then employed in 3D bioprinted liver models with bioink materials that closely mimic the natural extracellular matrix. The constructed humanized 3D bioprinted livers (h3DPLs) exhibited mature hepatocyte functions, including albumin expression, glycogen storage, and uptake/release of indocyanine green and acetylated low-density lipoprotein. Notably, h3DPLs demonstrated increased sensitivity to hepatotoxic agents such as acetaminophen (APAP), making them a promising platform for studying drug-induced liver injury. Furthermore, our model accurately reflected the impact of rifampin, a cytochrome P450 inducer, on CYP2E1 levels and APAP hepatotoxicity. These results highlight the potential of hiHep-based h3DPLs as a cost-effective and high-performance alternative for personalized liver toxicity screening and preclinical drug testing, paving the way for improved drug development strategies and personalized therapeutic interventions.
    Keywords:  3D bioprinting; Liver toxicity screening; human-induced hepatocytes; humanized 3D bioprinted livers
    DOI:  https://doi.org/10.1177/20417314241313341
  4. J Funct Biomater. 2025 Jan 16. pii: 28. [Epub ahead of print]16(1):
      The application of three-dimensional (3D) printing/bioprinting technologies and cell therapies has garnered significant attention due to their potential in the field of regenerative medicine. This paper aims to provide a comprehensive overview of 3D printing/bioprinting technology and cell therapies, highlighting their results in diverse medical applications, while also discussing the capabilities and limitations of their combined use. The synergistic combination of 3D printing and cellular therapies has been recognised as a promising and innovative approach, and it is expected that these technologies will progressively assume a crucial role in the treatment of various diseases and conditions in the foreseeable future. This review concludes with a forward-looking perspective on the future impact of these technologies, highlighting their potential to revolutionize regenerative medicine through enhanced tissue repair and organ replacement strategies.
    Keywords:  3D printing; additive manufacturing; biomaterials; biomedical applications; bioprinting; cellular therapies; tissue regeneration
    DOI:  https://doi.org/10.3390/jfb16010028
  5. Biomater Sci. 2025 Jan 21.
      Thrombosis, a major cause of morbidity and mortality worldwide, presents a complex challenge in cardiovascular medicine due to the intricacy of clotting mechanisms in living organisms. Traditional research approaches, including clinical studies and animal models, often yield conflicting results due to the inability to control variables in these complex systems, highlighting the need for more precise investigative tools. This review explores the evolution of in vitro thrombosis models, from conventional polydimethylsiloxane (PDMS)-based microfluidic devices to advanced hydrogel-based systems and cutting-edge 3D bioprinted vascular constructs. We discuss how these emerging technologies, particularly vessel-on-a-chip platforms, are enabling researchers to control previously unmanageable factors, thereby offering unprecedented opportunities to pinpoint specific clotting mechanisms. While PDMS-based devices offer optical transparency and fabrication ease, their inherent limitations, including non-physiological rigidity and surface properties, have driven the development of hydrogel-based systems that better mimic the extracellular matrix of blood vessels. The integration of microfluidics with biomimetic materials and tissue engineering approaches has led to the development of sophisticated models capable of simulating patient-specific vascular geometries, flow dynamics, and cellular interactions under highly controlled conditions. The advent of 3D bioprinting further enables the creation of complex, multi-layered vascular structures with precise spatial control over geometry and cellular composition. Despite significant progress, challenges remain in achieving long-term stability, incorporating immune components, and translating these models to clinical applications. By providing a comprehensive overview of current advancements and future prospects, this review aims to stimulate further innovation in thrombosis research and accelerate the development of more effective, personalized approaches to thrombosis prevention and treatment.
    DOI:  https://doi.org/10.1039/d4bm01354a
  6. Food Sci Anim Resour. 2025 Jan;45(1): 126-144
      Rapid population growth and a corresponding increase in the demand for animal-derived proteins have led to food supply challenges and the need for alternative and sustainable meat production methods. Therefore, this study explored the importance of cell engineering technology-based three-dimensional bioprinting and bioinks, which play key roles in cultured meat production. In cultured meat production, bioinks have a significant effect on cell growth, differentiation, and mechanical stability. Hence, in this study, the characteristics of animal-, plant-, and marine-based bioinks were compared and analyzed, and the impact of each bioink on cultured meat production was evaluated. In particular, animal-based bioinks have the potential to produce cultured meat that is similar to conventional meat and are considered the most suitable bioinks for commercialization. Although plant- and marine-based bioinks are ecofriendly and have fewer religious restrictions, they are limited in terms of mechanical stability and consumer acceptance. Therefore, further research is required to develop and apply optimal animal-based bioinks for commercialization of cultured meat, particularly to improve its mechanical compatibility.
    Keywords:  bioink; cell scaffold; meat culture; three-dimensional (3D) bioprint
    DOI:  https://doi.org/10.5851/kosfa.2024.e120
  7. Drug Metab Pharmacokinet. 2025 Jan 04. pii: S1347-4367(24)00055-7. [Epub ahead of print]60 101049
      Recently human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have become an attractive platform to evaluate drug responses for cardiotoxicity testing and disease modeling. Moreover, three-dimensional (3D) cardiac models, such as engineered heart tissues (EHTs) developed by bioengineering approaches, and cardiac spheroids (CSs) formed by spherical aggregation of hPSC-CMs, have been established as useful tools for drug discovery and transplantation. These 3D models overcome many of the shortcomings of conventional 2D hPSC-CMs, such as immaturity of the cells. Cardiac organoids (COs), like other organs, have also been studied to reproduce structures that resemble a heart in vivo more closely and optimize various culture conditions. Heart-on-a-chip (HoC) developed by a microfluidic chip-based technology that enables real-time monitoring of contraction and electrical activity, provides multifaceted information that is essential for capturing natural tissue development in vivo. Recently, 3D experimental systems have been developed to study organ interactions in vitro. This review aims to discuss the developments and advancements of hPSC-CMs and 3D cardiac tissues.
    Keywords:  Cardiac organoids; Cardiac spheroids; Drug discovery; Engineered heart tissue; Heart on a chip; Human pluripotent stem cells derived cardiomyocytes
    DOI:  https://doi.org/10.1016/j.dmpk.2024.101049
  8. Bioact Mater. 2025 Apr;46 365-385
      Articular cartilage has a limited self-healing capacity, leading to joint degeneration and osteoarthritis over time. Therefore, bioactive scaffolds are gaining attention as a promising approach to regenerating and repairing damaged articular cartilage through tissue engineering. In this study, we reported on a novel 3D bio-printed proteinaceous bioactive scaffolds combined with natural porcine cancellous bone dECM, tempo-oxidized cellulose nanofiber (TOCN), and alginate carriers for TGF-β1, FGF-18, and ADSCs to repair cartilage defects. The characterization results demonstrate that the 3D scaffolds are physically stable and facilitate a controlled dual release of TGF-β1 and FGF-18. Moreover, the key biological proteins within the bioactive scaffold actively interact with the biological systems to create a favorable microenvironment for cartilage regeneration. Importantly, the in vitro, in vivo, and in silico simulation showed that the scaffolds promote stem cell recruitment, migration, proliferation, and ECM deposition, and synergistic effects of TGF-β1/FGF-18 with the bioactive scaffolds significantly regulate stem cell chondrogenesis by activating the PI3K/AKT and TGFβ1/Smad4 signaling pathways. After implantation, the proteinaceous bioactive scaffold led to the regeneration of mechanically robust, full-thickness cartilage tissue that closely resembles native cartilage. Thus, these findings may provide a promising approach for regulating stem cell chondrogenesis and treating in situ cartilage regeneration.
    Keywords:  3D bioprinting; Cartilage regeneration; TGF-β1/FGF-18; TOCN; dECM
    DOI:  https://doi.org/10.1016/j.bioactmat.2024.12.021
  9. ACS Appl Mater Interfaces. 2025 Jan 24.
      Multidrug resistance (MDR) refers to the ability of cancer cells to resist various anticancer drugs and release them from the cells. This phenomenon is widely recognized as a significant barrier that must be overcome in chemotherapy. MDR varies depending on the number and expression level of the ATP-binding cassette transporter (ABC transporter), which is expressed differently in various cancer cells. Therefore, the dose of anticancer drugs should be adjusted according to the extent of MDR. The demand for drug screening that considers the differences in MDR is increasing in the process of drug discovery. In this study, three types of tumor spheroids were fabricated from HeLa (MRP1-/BCRP-), HepG2 (MRP1+/BCRP-), and A549 cells (MRP1+/BCRP+) using three-dimensional (3D) bioprinting. The fabricated tumor spheroids maintained their own MDR phenotypes. The EC50 values of doxorubicin (DOX) against the three tumor spheroids were more than 2-fold higher than those against the 2D cells. In addition, the EC50 value of DOX against tumor spheroids was proportional to the number of ABC transporters. The EC50 value of DOX against A549 tumor spheroids had the largest value of 9.5 μM among the three spheroids. In addition, the EC50 values of DOX against HepG2 and A549 tumor spheroids were remarkably reduced when they were treated with ABC transporter inhibitors, such as MK-571 against MRP1 and/or NOV against BCRP. These results demonstrate the successful construction of a 3D bioprinting-based screening platform to quantitatively evaluate the anticancer efficacy of chemodrugs, considering the MDR of cancer cells.
    Keywords:  3D bioprinting; ABC transporter; anticancer treatment; half-maximal effective concentration; multidrug resistance
    DOI:  https://doi.org/10.1021/acsami.4c19291
  10. Expert Rev Gastroenterol Hepatol. 2025 Jan 20. 1-6
       INTRODUCTION: Liver fibrosis, marked by excessive extracellular matrix deposition, is a significant consequence of chronic liver injuries from various conditions. It can progress to end-stage liver disease, with liver transplantation often being the only treatment option. Recent advancements in 3D-organoid technology have transformed liver disease research by providing models that mimic the human liver's physiological environment, offering insights into mechanisms of fibrosis and potential therapies.
    AREAS COVERED: This report highlights cellular and molecular factors leading to liver fibrosis and the limitations of 2D in vitro models in replicating complex liver dynamics. It emphasizes the advantages of 3D-liver organoids as promising tools for advancing research and drug discovery, providing greater accuracy than traditional models. Additionally, it discusses recent advancements in the development and future applications of liver organoids in fibrosis research.
    EXPERT OPINION: Liver organoids currently lack cellular diversity and essential features such as vascular, neuronal, microbiome, and immune responses, limiting their effectiveness in mature fibrosis models. Addressing these shortcomings through bioengineering advancements and emerging technologies like CRISPR/Cas9 will enhance the utility of liver organoids.
    Keywords:  3D cultures; drug discovery; inflammation; liver fibrosis; organoids
    DOI:  https://doi.org/10.1080/17474124.2025.2455581