bims-biprem Biomed News
on Bioprinting for regenerative medicine
Issue of 2024‒05‒05
twelve papers selected by
Seerat Maqsood, University of Teramo



  1. Arch Dermatol Res. 2024 May 02. 316(5): 147
      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.
    Keywords:  Mohs micrographic surgery; Skin bioprinting; Skin regeneration; Three-dimensional bioprinting
    DOI:  https://doi.org/10.1007/s00403-024-02893-6
  2. Acta Biomater. 2024 Apr 30. pii: S1742-7061(24)00221-6. [Epub ahead of print]
      Skeletal muscle is a pro-regenerative tissue, that utilizes a tissue-resident stem cell system to effect repair upon injury. Despite the demonstrated efficiency of this system in restoring muscle mass after many acute injuries, in conditions of severe trauma such as those evident in volumetric muscle loss (VML) (>20% by mass), this self-repair capability is unable to restore tissue architecture, requiring interventions which currently are largely surgical. As a possible alternative, the generation of artificial muscle using tissue engineering approaches may also be of importance in the treatment of VML and muscle diseases such as dystrophies. Three-dimensional (3D) bioprinting has been identified as a promising technique for regeneration of the complex architecture of skeletal muscle. This review discusses existing treatment strategies following muscle damage, recent progress in bioprinting techniques, the bioinks used for muscle regeneration, the immunogenicity of scaffold materials, and in vitro and in vivo maturation techniques for 3D bio-printed muscle constructs. The pros and cons of these bioink formulations are also highlighted. Finally, we present the current limitations and challenges in the field and critical factors to consider for bioprinting approaches to become more translational and to produce clinically relevant engineered muscle. STATEMENT OF SIGNIFICANCE: This review discusses the physiopathology of muscle injuries and existing clinical treatment strategies for muscle damage, the types of bioprinting techniques that have been applied to bioprinting of muscle, and the bioinks commonly used for muscle regeneration. The pros and cons of these bioinks are highlighted. We present a discussion of existing gaps in the literature and critical factors to consider for the translation of bioprinting approaches and to produce clinically relevant engineered muscle. Finally, we provide insights into what we believe will be the next steps required before the realization of the application of tissue-engineered muscle in humans. We believe this manuscript is an insightful, timely, and instructive review that will guide future muscle bioprinting research from a fundamental construct creation approach, down a translational pathway to achieve the desired impact in the clinic.
    Keywords:  Bioprinting; Hydrogels; Muscle; Regeneration
    DOI:  https://doi.org/10.1016/j.actbio.2024.04.038
  3. Macromol Biosci. 2024 Apr 29. e2400035
      3D bioprinting allows rapid automated fabrication and can be applied for high throughput generation of biomimetic constructs for in vitro drug screening. Decellularized extracellular matrix (dECM) hydrogel is a popular biomaterial choice for tissue engineering and studying carcinogenesis as a tumor microenvironmental mimetic. This study proposes a method for high throughput bioprinting with decellularized adipose tissue (DAT) based hydrogels for 3D breast cancer modeling. A comparative analysis of decellularization protocol using detergent-based and detergent-free decellularization methods for caprine-origin adipose tissue is performed, and the efficacy of dECM hydrogel for 3D cancer modeling is assessed. Histological, biochemical, morphological, and biological characterization and analysis showcase the cytocompatibility of DAT hydrogel. The rheological property of DAT hydrogel and printing process optimization is assessed to select a bioprinting window to attain 3D breast cancer models. The bioprinted tissues are characterized for cellular viability and tumor cell-matrix interactions. Additionally, an approach for breast cancer modeling is shown by performing rapid high throughput bioprinting in a 96-well plate format, and in vitro drug screening using 5-fluorouracil is performed on 3D bioprinted microtumors. The results of this study suggest that high throughput bioprinting of cancer models can potentially have downstream clinical applications like multi-drug screening platforms and personalized disease models.
    Keywords:  3D bioprinting; adipose tissue; breast cancer; decellularized extracellular matrix hydrogel; high throughput
    DOI:  https://doi.org/10.1002/mabi.202400035
  4. Biomed Mater. 2024 May 02.
      Porous tantalum scaffolds offer a high degree of biocompatibility and have a low friction coefficient. In addition, their biomimetic porous structure and mechanical properties, which closely resemble human bone tissue, make them a popular area of research in the field of bone defect repair. With the rapid advancement of additive manufacturing, 3D-printed porous tantalum scaffolds have increasingly emerged in recent years, offering exceptional design flexibility, as well as facilitating the fabrication of intricate geometries and complex pore structures that similar to human anatomy. This review provides a comprehensive description of the techniques, procedures, and specific parameters involved in the 3D printing of porous tantalum scaffolds. Concurrently, the review provides a summary of the mechanical properties, osteogenesis and antibacterial properties of porous tantalum scaffolds. The use of surface modification techniques and the drug carriers can enhance the characteristics of porous tantalum scaffolds. Accordingly, the review discusses the application of these porous tantalum materials in clinical settings. Multiple studies have demonstrated that 3D-printed porous tantalum scaffolds exhibit exceptional corrosion resistance, biocompatibility, and osteogenic properties. As a result, they are considered highly suitable biomaterials for repairing bone defects. Despite the rapid development of 3D-printed porous tantalum scaffolds, they still encounter challenges and issues when used as bone defect implants in clinical applications. Ultimately, a concise overview of the primary challenges faced by 3D-printed porous tantalum scaffolds is offered, and corresponding insights to promote further exploration and advancement in this domain are presented.
    Keywords:  3D printing; Bone reconstruction; Mechanical properties; Multi -biological functions; Porous tantalum scaffolds
    DOI:  https://doi.org/10.1088/1748-605X/ad46d2
  5. Int J Biol Macromol. 2024 Apr 25. pii: S0141-8130(24)02525-X. [Epub ahead of print] 131720
      The human nervous system is an incredibly intricate physiological network, and neural cells lack the ability to repair and regenerate after a brain injury. 3-dimensional (3D) bioprinting technology offers a promising strategy for constructing biomimetic organ constructs and in vitro brain/disease models. The bioink serves as a pivotal component that emulates the microenvironment of biomimetic construct and exerts a profound influence on cellular behaviors. In this study, a series of mechanically adjustable and dual crosslinking bioinks were developed using photocrosslinkable methacrylated silk fibroin (SilMA) in combination with the ionic crosslinking material, pectin, or pectin methacryloyl (PecMA) with silk fibroin (SF) supplementation. SilMA/pectin exhibited superior properties, with SilMA providing biocompatibility and adjustable mechanical properties, while the addition of pectin enhanced printability. The porous structure supported neural cell growth, and 15 % SilMA/0.5 % pectin bioinks displayed excellent printability and shape fidelity. Neural stem/progenitor cells (NSPCs)-loaded bioinks were used to construct a 3D brain model, demonstrating sustained vitality and high neuronal differentiation without the need for growth factors. The SilMA/pectin bioinks demonstrated adjustable mechanical properties, favorable biocompatibility, and an environment highly conducive to neural induction, offering an alternative approach for neural tissue engineering applications or in vitro brain models.
    Keywords:  3D bioprinting; Bioink; In vitro brain model; Neural stem/progenitor cells (NSPCs); Pectin; Silk fibroin/methacrylated silk fibroin (SilMA)
    DOI:  https://doi.org/10.1016/j.ijbiomac.2024.131720
  6. BMC Biotechnol. 2024 Apr 30. 24(1): 25
      The reconstruction of a stable, nipple-shaped cartilage graft that precisely matches the natural nipple in shape and size on the contralateral side is a clinical challenge. While 3D printing technology can efficiently and accurately manufacture customized complex structures, it faces limitations due to inadequate blood supply, which hampers the stability of nipple-shaped cartilage grafts produced using this technology. To address this issue, we employed a biodegradable biomaterial, Poly(lactic-co-glycolic acid) (PLGA), loaded with Cell-Free Fat Extract (Ceffe). Ceffe has demonstrated the ability to promote angiogenesis and cell proliferation, making it an ideal bio-ink for bioprinting precise nipple-shaped cartilage grafts. We utilized the Ceffe/PLGA scaffold to create a porous structure with a precise nipple shape. This scaffold exhibited favorable porosity and pore size, ensuring stable shape maintenance and satisfactory biomechanical properties. Importantly, it could release Ceffe in a sustained manner. Our in vitro results confirmed the scaffold's good biocompatibility and its ability to promote angiogenesis, as evidenced by supporting chondrocyte proliferation and endothelial cell migration and tube formation. Furthermore, after 8 weeks of in vivo culture, the Ceffe/PLGA scaffold seeded with chondrocytes regenerated into a cartilage support structure with a precise nipple shape. Compared to the pure PLGA group, the Ceffe/PLGA scaffold showed remarkable vascular formation, highlighting the beneficial effects of Ceffe. These findings suggest that our designed Ceffe/PLGA scaffold with a nipple shape represents a promising strategy for precise nipple-shaped cartilage regeneration, laying a foundation for subsequent nipple reconstruction.
    Keywords:  3D printing; Ceffe; Nipple reconstruction; Tissue engineering; Vascularization
    DOI:  https://doi.org/10.1186/s12896-024-00848-3
  7. Am J Orthod Dentofacial Orthop. 2024 Apr 26. pii: S0889-5406(24)00137-9. [Epub ahead of print]
      INTRODUCTION: The purpose of this study was to examine the use of orthodontic 3-dimensional (3D) printing technology in North America and to understand why orthodontists are, or are not, incorporating 3D printing technology in their practices.METHODS: A survey questionnaire was delivered on a secure online platform, RedCap (Case Western Reserve University Clinical and Translational Science Award; no. UL1TR002548). The survey consisted of 14-34 items with branching logic. The association between participant demographics and in-house 3D printing was assessed using a chi-square test of independence.
    RESULTS: A total of 518 responses were recorded. The highest number of responses came from respondents in the 36-45-year age group. Most of the respondents were practice owners; 46.9% had 3D printers in their office. Chi-square tests of independence were performed on the data to see which associations existed. The strongest statistical associations with using an in-house 3D printer are seen with patient load, practice type, years since residency, and orthodontist's position.
    CONCLUSIONS: Approximately 75% of orthodontists use 3D printing technology in some capacity in North America. Major factors that influenced orthodontists to incorporate 3D printing technology into their office were self-interest and research. Major factors that have prevented orthodontists from not incorporating 3D printing technology into their office were space for equipment/ventilation and digital workflow training deficit. Orthodontists use their 3D printers mostly to make plastic retainers from printed models. The strongest associations with using in-house 3D printers are seen in patient load, practice type, years since residency, and orthodontist position. Increasing patient load and being in private practice increases the likelihood of having a 3D printer.
    DOI:  https://doi.org/10.1016/j.ajodo.2024.03.014
  8. Int J Biol Macromol. 2024 Apr 26. pii: S0141-8130(24)02646-1. [Epub ahead of print]268(Pt 2): 131841
      BACKGROUND: Targeted nanoparticles (NPs) are aimed at improving clinical outcomes by enhancing the diagnostic and therapeutic efficacy of drugs in the treatment of Alzheimer's disease (AD).METHODS: Curcumin (CUR)-loaded poly-lactic-co-glycolic acid (PLGA) NPs (CNPs) were produced to demonstrate a prolonged release and successfully embedded into 3D printed sodium alginate (SA)/gelatin (GEL) scaffolds that can dissolve rapidly sublingually. Characterization and in vitro activity of the NPs and scaffolds were evaluated.
    RESULTS: Based on the in vitro drug release studies, 99.6 % of the encapsulated CUR was released in a controlled manner within 18 days for the CNPs. In vitro cell culture studies showed that all samples exhibited cell viability above 84.2 % and no significant cytotoxic effect on SH-SY5Y cells. The samples were analyzed through 2 different pathways by PCR analysis. Real-time PCR results indicated that CNP and CNP-embedded SA/GEL scaffolds (CNPSGS) may show neuroprotective effects by modulating the Wnt/β-catenin pathway. The gene expression level of β-catenin slightly increased compared to the gene expression levels of other proteins and enzymes with these treatments. However, the PI3K/Akt/GSK-3β signaling pathway was regulated at the same time because of the crosstalk between these 2 pathways.
    CONCLUSION: CNPSGS might be an effective therapeutic alternative for AD treatment.
    Keywords:  3D printing; Alzheimer's disease; Curcumin
    DOI:  https://doi.org/10.1016/j.ijbiomac.2024.131841
  9. Atherosclerosis. 2024 Apr 15. pii: S0021-9150(24)00104-7. [Epub ahead of print] 117544
      Steatotic liver disease (SLD) refers to a spectrum of diseases caused by hepatic lipid accumulation. SLD has emerged as the leading cause of chronic liver disease worldwide. Despite this burden and many years, understanding the pathophysiology of this disease is challenging due to the inaccessibility to human liver specimens. Therefore, cell-based in vitro systems are widely used as models to investigate the pathophysiology of SLD. Culturing hepatic cells in monolayers causes the loss of their hepatocyte-specific phenotype and, consequently, tissue-specific function and architecture. Hence, three-dimensional (3D) culture models allow cells to mimic the in vivo microenvironment and spatial organization of the liver unit. The utilization of 3D in vitro models minimizes the drawbacks of two-dimensional (2D) cultures and aligns with the 3Rs principles to alleviate the number of in vivo experiments. This article provides an overview of liver 3D models highlighting advantages and limitations, and culminates by discussing their applications in pharmaceutical and biomedical research.
    Keywords:  Buds; MAFLD; MASH; MASLD; NASH; Organoids; Spheroids
    DOI:  https://doi.org/10.1016/j.atherosclerosis.2024.117544
  10. Nihon Yakurigaku Zasshi. 2024 ;159(3): 144-149
      We have been making 3D tissues consist of cells only, based on the corporate philosophy of "contributing to dramatic advances in medical care through the practical application of innovative 3D cell stacking technology." Currently, in the field of regenerative medicine, we are working toward obtaining approval from the Ministry of Health, Labor and Welfare and commercializing large artificial organs that are made from patients' own cells and have functions such as nerve regeneration, osteochondral regeneration, and blood vessels. On the other hand, this three-dimensional cell stacking technology can be extended to technology for culturing cells in an environment similar to the human body, and is expected to serve as a new methodology for evaluating the effects of new products in various fields on living organisms. Therefore, we are planning a business to provide developers of pharmaceuticals, foods, cosmetics, etc. with a small device called "Functional Cell Device (FCD)" that reproduces some of the functions of human organs outside the body. As the first step, we have developed a three-dimensional liver construct (3D mini-liver). The in vitro human liver model has a wide range of usage, such as evaluation of hepatotoxicity of drugs, elucidation of drug metabolism mechanism, and model of liver disease. In this report, we will outline it together with actual examples in regenerative medicine.
    DOI:  https://doi.org/10.1254/fpj.23049
  11. J Mater Chem B. 2024 Apr 30.
      The application of three- and four-dimensional (3D/4D) printing in cancer research represents a significant advancement in understanding and addressing the complexities of cancer biology. 3D/4D materials provide more physiologically relevant environments compared to traditional two-dimensional models, allowing for a more accurate representation of the tumor microenvironment that enables researchers to study tumor progression, drug responses, and interactions with surrounding tissues under conditions similar to in vivo conditions. The dynamic nature of 4D materials introduces the element of time, allowing for the observation of temporal changes in cancer behavior and response to therapeutic interventions. The use of 3D/4D printing in cancer research holds great promise for advancing our understanding of the disease and improving the translation of preclinical findings to clinical applications. Accordingly, this review aims to briefly discuss 3D and 4D printing and their advantages and limitations in the field of cancer. Moreover, new techniques such as 5D/6D printing and artificial intelligence (AI) are also introduced as methods that could be used to overcome the limitations of 3D/4D printing and opened promising ways for the fast and precise diagnosis and treatment of cancer.
    DOI:  https://doi.org/10.1039/d4tb00310a
  12. Methods Mol Biol. 2024 ;2803 35-48
      The lack of a precise noninvasive, clinical evaluation method for cardiac fibrosis hinders the development of successful treatments that can effectively work in physiological settings, where tissues and organs are interconnected and moderating drug responses. To address this challenge and advance personalized medicine, researchers have turned to human-induced pluripotent stem (iPS) cells, which can be differentiated to resemble the human heart in terms of structure, function and cellular composition. In this chapter, we present an assay protocol that uses these iPS cells to generate heart organoids for the in vitro evaluation of cardiac fibrosis. By establishing this biological platform, we pave the way for conducting phenotype evaluation and treatment screening in a multiscale approach, aiming to discover effective interventions for the treatment of cardiac fibrosis.
    Keywords:  Cardiac fibrosis; Cardiac organoid; Differentiation; Fibroblasts; Heart organoid; Reactive fibrosis; Replacement fibrosis; TGFβ; hiPSCs
    DOI:  https://doi.org/10.1007/978-1-0716-3846-0_3